Ninh T. Nguyen Stacy A. Brethauer John M. Morton Jaime Ponce Raul J. Rosenthal Editors The ASMBS Textbook of Bariatric Surgery Second Edition 123 The ASMBS Textbook of Bariatric Surgery Ninh T. Nguyen • Stacy A. Brethauer John M. Morton • Jaime Ponce Raul J. Rosenthal Editors The ASMBS Textbook of Bariatric Surgery Second Edition Editors Ninh T. Nguyen Department of Surgery, Division of Gastrointestinal Surgery University of California, Irvine Medical Center Orange, CA USA John M. Morton Bariatric and Minimally Invasive Surgery Division Yale School of Medicine New Haven, CT USA Stacy A. Brethauer Department of Surgery The Ohio State University Columbus, OH USA Jaime Ponce Bariatric Surgery, Metabolic and Bariatric Care CHI Memorial Hospital Chattanooga, TN USA Raul J. Rosenthal Department of General Surgery, The Bariatric and Metabolic Institute Cleveland Clinic Florida Weston, FL USA ISBN 978-3-030-27020-9 ISBN 978-3-030-27021-6 https://doi.org/10.1007/978-3-030-27021-6 (eBook) © Springer Nature Switzerland AG 2020 This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or part of the material is concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way, and transmission or information storage and retrieval, electronic adaptation, computer software, or by similar or dissimilar methodology now known or hereafter developed. The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication does not imply, even in the absence of a specific statement, that such names are exempt from the relevant protective laws and regulations and therefore free for general use. The publisher, the authors, and the editors are safe to assume that the advice and information in this book are believed to be true and accurate at the date of publication. Neither the publisher nor the authors or the editors give a warranty, expressed or implied, with respect to the material contained herein or for any errors or omissions that may have been made. The publisher remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. This Springer imprint is published by the registered company Springer Nature Switzerland AG The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland Preface The American Society for Metabolic and Bariatric Surgery (ASMBS) is comprised of a dynamic group of surgeons, physicians, and integrated health members, all of whom are constantly challenged to improve the care of patients with obesity. As acknowledged in a landmark 2013 decision by the American Medical Association, clinically severe obesity is a disease process that is associated with multiple life-threatening conditions that may lead to premature death. As repeatedly and consistently demonstrated by literature evidence, metabolic and bariatric surgery has shown to be the only long-lasting effective treatment for obesity and its related comorbidities. The field of bariatric surgery has changed tremendously over that past three decades since the ASMBS’s founding in 1983. Application of minimally invasive surgery had revolutionized the way we perform modern-day bariatric surgery. Once a large midline incision has changed to multiple small surgical incisions with enhanced recovery, shortened length of hospitalization, and reduced morbidity and mortality. In the late 1990s, less than 12,000 bariatric operations were performed yearly in the United States, with high rates of morbidity and mortality. This number of operations has increased exponentially over the subsequent years, and currently more than 225,000 operations are being performed annually. The initial growth directly correlates with the development and transition from the open to minimally invasive bariatric surgery, and by 2005, the number of laparoscopic Roux-en-Y gastric bypass cases performed in the United States surpassed that of the open Roux-en-Y gastric bypass. Since 2010, the laparoscopic sleeve gastrectomy has proven to be a safe and effective bariatric operation and is now the most commonly performed bariatric operation in the Unites States and worldwide. Along with advancement in surgical technique, the quality of bariatric surgery had also improved with implementation of accredited centers designated by the ASMBS and the American College of Surgeons. This dynamic field of bariatric surgery will continue to grow with enhanced understanding of the mechanisms of action of our bariatric operations and offering of other minimally invasive approaches including endoscopic bariatric procedures. As the needs of the society and its members evolve, the ASMBS continues its commitment to serve the educational needs of our members. Our annual meeting is the primary venue to disseminate new information and educational materials to clinical professionals. To enhance and augment these educational offerings, we published the comprehensive ASMBS textbook of bariatric surgery, first edition in 2014. The development of this book reflects the commitment of the ASMBS leadership’s goal of providing the most up-to-date education for our members. Designed to be the most inclusive textbook on the topic of bariatric surgery and integrated health services, the textbook is comprised of two volumes. The first volume is devoted to the science and practices of bariatric surgery, while the second volume focuses on the medical, psychological, and nutritional management of the bariatric patients. Since 2014, the practice of bariatric surgery has continued to change at a rapid pace, and we are excited to provide you with the second edition of the textbook on the science and practices of bariatric surgery and obesity management. This second edition has expanded from 5 to 8 sections to include new sections on quality in bariatric surgery, endoscopic bariatric surgery, and management of bariatric complications with expansion from 43 to 56 chapters. v vi Preface Similar to the first edition, each chapter in this book was written by a world-renowned expert in the field. A comprehensive textbook that adheres to the highest standards is a major undertaking, and we, the editors, are grateful and indebted to every author who has devoted time and effort to research the most important evidence-based information and report it in a concise and easy-to-read chapter. We believe that this second edition of the ASMBS Textbook of Bariatric Surgery will continue to be the leading source of scientific information for surgeons, physicians, residents, students, and integrated health members today and for years to come. We hope you enjoy the textbook and would love to hear your comments. Orange, CA, USA Columbus, OH, USA New Haven, CT, USA Chattanooga, TN, USA Weston, FL, USA Falls Church, VA, USA Philadelphia, PA, USA Danville, PA, USA Ninh T. Nguyen, MD Stacy A. Brethauer, MD John M. Morton, MD, MPH Jaime Ponce, MD Raul J. Rosenthal, MD Jeanne Blankenship, RD David Sarwer, PhD Christopher Still, MD Contents Part I Basic Considerations 1Epidemiology and Discrimination in Obesity���������������������������������������������������������� 3 R. Armour Forse, Monica M. Betancourt-Garcia, and Michelle Cordoba Kissee Introduction����������������������������������������������������������������������������������������������������������������� 3 Definition of Obesity��������������������������������������������������������������������������������������������������� 3 Epidemiology of Obesity ������������������������������������������������������������������������������������������� 5 Racial, Ethnic, and Income Disparities����������������������������������������������������������������������� 8 Trends in Obesity ������������������������������������������������������������������������������������������������������� 9 Discrimination in Obesity������������������������������������������������������������������������������������������� 10 Conclusion ����������������������������������������������������������������������������������������������������������������� 13 Question Section��������������������������������������������������������������������������������������������������������� 13 References������������������������������������������������������������������������������������������������������������������� 13 2The Pathophysiology of Obesity and Obesity-Related Disease������������������������������� 15 Robert W. O’Rourke Introduction����������������������������������������������������������������������������������������������������������������� 16 Pathophysiology of Obesity ��������������������������������������������������������������������������������������� 17 Pathophysiology of Obesity-Related Disease������������������������������������������������������������� 25 Conclusion ����������������������������������������������������������������������������������������������������������������� 33 Question Section��������������������������������������������������������������������������������������������������������� 33 References������������������������������������������������������������������������������������������������������������������� 34 3History of the Development of Metabolic/Bariatric Surgery��������������������������������� 37 Elias Chousleb, Jaime A. Rodriguez, and J. Patrick O’Leary History of Bariatric Surgery��������������������������������������������������������������������������������������� 37 Metabolic ������������������������������������������������������������������������������������������������������������������� 42 Minimally Invasive Techniques ��������������������������������������������������������������������������������� 43 Hormonal Weight Loss����������������������������������������������������������������������������������������������� 44 Accreditation��������������������������������������������������������������������������������������������������������������� 45 Conclusion ����������������������������������������������������������������������������������������������������������������� 45 References������������������������������������������������������������������������������������������������������������������� 45 4The History of the American Society for Metabolic and Bariatric Surgery��������� 47 Robin P. Blackstone Introduction����������������������������������������������������������������������������������������������������������������� 47 The Era of Inquiry 1967–1988����������������������������������������������������������������������������������� 47 The Era of Rapid Growth 1989–2004������������������������������������������������������������������������� 50 Integrated Health��������������������������������������������������������������������������������������������������������� 51 Growth of the Society������������������������������������������������������������������������������������������������� 52 Medical Liability��������������������������������������������������������������������������������������������������������� 53 The Era of Quality and Engagement from 2004 to Present ��������������������������������������� 53 International Affiliations��������������������������������������������������������������������������������������������� 58 vii viii Contents The ASMBS Foundation��������������������������������������������������������������������������������������������� 58 Obesity Week ������������������������������������������������������������������������������������������������������������� 58 Conclusion ����������������������������������������������������������������������������������������������������������������� 59 References������������������������������������������������������������������������������������������������������������������� 59 5Physiological Mechanisms of Bariatric Procedures������������������������������������������������� 61 David Romero Funes, Emanuele Lo Menzo, Samuel Szomstein, and Raul J. Rosenthal Introduction����������������������������������������������������������������������������������������������������������������� 61 Mechanism of Action������������������������������������������������������������������������������������������������� 62 Malabsorption������������������������������������������������������������������������������������������������������������� 62 Caloric Restriction ����������������������������������������������������������������������������������������������������� 62 Energy Expenditure���������������������������������������������������������������������������������������������������� 63 Changes in Eating Behavior��������������������������������������������������������������������������������������� 63 Entero-Hormones, Incretins, and Intestinal Adaptation��������������������������������������������� 63 Mechanisms of Diabetes Resolution��������������������������������������������������������������������������� 67 Neuroendocrine Mechanism��������������������������������������������������������������������������������������� 67 Vagus Nerve ��������������������������������������������������������������������������������������������������������������� 67 Bile Acids (BA)����������������������������������������������������������������������������������������������������������� 68 Gastrointestinal Microflora����������������������������������������������������������������������������������������� 68 Adipose Tissue ����������������������������������������������������������������������������������������������������������� 69 Leptin ������������������������������������������������������������������������������������������������������������������������� 69 Adiponectin����������������������������������������������������������������������������������������������������������������� 70 β(Beta)-Cell Changes������������������������������������������������������������������������������������������������� 70 End-Organ Changes ��������������������������������������������������������������������������������������������������� 70 Gastrointestinal-Renal Axis ��������������������������������������������������������������������������������������� 71 Bariatric Surgery and the Control of Blood Pressure Through the GI-Renal Axis����� 71 Conclusion ����������������������������������������������������������������������������������������������������������������� 72 Question Section��������������������������������������������������������������������������������������������������������� 72 References������������������������������������������������������������������������������������������������������������������� 72 6Indications and Contraindications for Bariatric Surgery��������������������������������������� 77 Christopher DuCoin, Rachel L. Moore, and David A. Provost Introduction����������������������������������������������������������������������������������������������������������������� 77 Indications for Metabolic and Bariatric Surgery�������������������������������������������������������� 77 Specific Considerations����������������������������������������������������������������������������������������������� 79 Conclusion ����������������������������������������������������������������������������������������������������������������� 80 Question Section��������������������������������������������������������������������������������������������������������� 80 References������������������������������������������������������������������������������������������������������������������� 80 7Preoperative Care of the Bariatric Patient��������������������������������������������������������������� 83 Renée M. Tholey and David S. Tichansky Introduction����������������������������������������������������������������������������������������������������������������� 83 Patient Selection��������������������������������������������������������������������������������������������������������� 83 Cardiac Evaluation����������������������������������������������������������������������������������������������������� 84 Preoperative VTE Evaluation������������������������������������������������������������������������������������� 84 Sleep Apnea and Obesity Hypoventilation Evaluation����������������������������������������������� 85 Evaluation of Upper Gastrointestinal Anatomy ��������������������������������������������������������� 86 Psychological Evaluation������������������������������������������������������������������������������������������� 86 Informed Consent������������������������������������������������������������������������������������������������������� 87 Conclusion: Standardized Care Pathways������������������������������������������������������������������ 87 Question Section��������������������������������������������������������������������������������������������������������� 87 References������������������������������������������������������������������������������������������������������������������� 88 Contents ix 8Anesthetic Considerations����������������������������������������������������������������������������������������� 89 Hendrikus J. M. Lemmens, John M. Morton, Cindy M. Ku, and Stephanie B. Jones Introduction����������������������������������������������������������������������������������������������������������������� 89 Preoperative Evaluation ��������������������������������������������������������������������������������������������� 89 Respiratory Issues Relevant to Anesthesia Management������������������������������������������� 89 Cardiovascular Issues Relevant to Anesthesia Management ������������������������������������� 90 Pharmacological Considerations��������������������������������������������������������������������������������� 91 Induction Agents��������������������������������������������������������������������������������������������������������� 91 Inhaled Anesthetics����������������������������������������������������������������������������������������������������� 93 Neuromuscular Blocking Agents ������������������������������������������������������������������������������� 94 Reversal Agents of Neuromuscular Blockade ����������������������������������������������������������� 95 Monitoring ����������������������������������������������������������������������������������������������������������������� 95 Preoperative Sedation������������������������������������������������������������������������������������������������� 96 Airway Management��������������������������������������������������������������������������������������������������� 96 Aspiration Risk����������������������������������������������������������������������������������������������������������� 97 Pneumoperitoneum: Implications for Ventilation, Hemodynamics, and Urine Output��������������������������������������������������������������������������������������������������������� 97 Fluid Management ����������������������������������������������������������������������������������������������������� 98 Postoperative Considerations������������������������������������������������������������������������������������� 98 Postoperative Nausea and Vomiting��������������������������������������������������������������������������� 99 Postoperative Analgesia ��������������������������������������������������������������������������������������������� 99 Question Section��������������������������������������������������������������������������������������������������������� 100 References������������������������������������������������������������������������������������������������������������������� 100 9Components of a Metabolic and Bariatric Surgery Center ����������������������������������� 103 Wayne J. English, D. Brandon Williams, and Aaron Bolduc Introduction����������������������������������������������������������������������������������������������������������������� 103 The Metabolic and Bariatric Surgery Accreditation and Quality Improvement Program����������������������������������������������������������������������������������� 103 Essential Components of a Metabolic and Bariatric Surgery (MBS) Center������������� 104 Specialty Consultants and Preoperative Clearances��������������������������������������������������� 111 Electronic and Remote Access to the Metabolic and Bariatric Surgery Center��������� 112 Metabolic and Bariatric Surgery in Adolescents�������������������������������������������������������� 112 Common Deficiencies Encountered During the MBSAQIP Site Survey������������������� 114 Conclusion ����������������������������������������������������������������������������������������������������������������� 115 Question Section��������������������������������������������������������������������������������������������������������� 115 References������������������������������������������������������������������������������������������������������������������� 116 10Evaluation of Preoperative Weight Loss������������������������������������������������������������������� 117 Hussna Wakily and Aurora D. Pryor Introduction����������������������������������������������������������������������������������������������������������������� 117 Principle Behind the Support of Preoperative Weight Loss��������������������������������������� 117 Review of the Data Supporting and Refuting the Benefit of Preoperative Weight Loss���������������������������������������������������������������������������������������������������������������� 118 Additional Considerations with PWL������������������������������������������������������������������������� 121 Study Limitations������������������������������������������������������������������������������������������������������� 121 ASMBS Position Statement��������������������������������������������������������������������������������������� 121 Conclusion ����������������������������������������������������������������������������������������������������������������� 122 Question Section��������������������������������������������������������������������������������������������������������� 122 References������������������������������������������������������������������������������������������������������������������� 122 x 11ASMBS Position Statements ������������������������������������������������������������������������������������� 123 Stacy A. Brethauer and Xiaoxi (Chelsea) Feng Introduction����������������������������������������������������������������������������������������������������������������� 123 Summary of Current Position Statements������������������������������������������������������������������� 123 Question Section��������������������������������������������������������������������������������������������������������� 133 References������������������������������������������������������������������������������������������������������������������� 134 Part II Primary Bariatric Surgery and Management of Complications 12Laparoscopic Gastric Bypass: Technique and Outcomes��������������������������������������� 139 Kelvin D. Higa and Pearl K. Ma Introduction����������������������������������������������������������������������������������������������������������������� 139 Preparation of the Patient������������������������������������������������������������������������������������������� 140 Positioning and Trocar Placement ����������������������������������������������������������������������������� 140 The Components��������������������������������������������������������������������������������������������������������� 142 Routing of the Roux Limb and Closure of Mesenteric Defects��������������������������������� 144 Outcomes ������������������������������������������������������������������������������������������������������������������� 144 Conclusion ����������������������������������������������������������������������������������������������������������������� 146 Question Section��������������������������������������������������������������������������������������������������������� 146 References������������������������������������������������������������������������������������������������������������������� 146 13Laparoscopic Sleeve Gastrectomy: Technique and Outcomes������������������������������� 149 Natan Zundel, Juan D. Hernandez R., and Michel Gagner Introduction����������������������������������������������������������������������������������������������������������������� 149 Preparation and Surgical Technique��������������������������������������������������������������������������� 150 Postoperative Period��������������������������������������������������������������������������������������������������� 152 Results������������������������������������������������������������������������������������������������������������������������� 152 Sleeve Gastrectomy or Gastric Bypass?��������������������������������������������������������������������� 156 Discussion������������������������������������������������������������������������������������������������������������������� 157 Conclusions����������������������������������������������������������������������������������������������������������������� 157 Question Section��������������������������������������������������������������������������������������������������������� 157 References������������������������������������������������������������������������������������������������������������������� 158 14Biliopancreatic Diversion with Duodenal Switch: Technique and Outcomes������� 161 Ranjan Sudan Introduction����������������������������������������������������������������������������������������������������������������� 161 Preoperative Assessment��������������������������������������������������������������������������������������������� 161 Technical Details��������������������������������������������������������������������������������������������������������� 162 Postoperative Management����������������������������������������������������������������������������������������� 164 Complications������������������������������������������������������������������������������������������������������������� 165 Outcomes ������������������������������������������������������������������������������������������������������������������� 166 Conclusion ����������������������������������������������������������������������������������������������������������������� 166 Question Section��������������������������������������������������������������������������������������������������������� 167 References������������������������������������������������������������������������������������������������������������������� 167 15Single Anastomosis Duodeno-ileostomy������������������������������������������������������������������� 169 Amit Surve, Daniel Cottam, Hinali Zaveri, and Samuel Cottam History������������������������������������������������������������������������������������������������������������������������� 169 Surgical Technique����������������������������������������������������������������������������������������������������� 170 Postoperative Care ����������������������������������������������������������������������������������������������������� 172 Weight Loss Outcomes with SADI-S������������������������������������������������������������������������� 172 Complications������������������������������������������������������������������������������������������������������������� 173 Rare Complications and Their Management ������������������������������������������������������������� 175 Nutritional Outcomes������������������������������������������������������������������������������������������������� 175 Contents Contents xi T2DM Resolution������������������������������������������������������������������������������������������������������� 175 SADI as Revision Procedure�������������������������������������������������������������������������������������� 177 Review of Literature��������������������������������������������������������������������������������������������������� 177 Conclusion ����������������������������������������������������������������������������������������������������������������� 177 Question Section��������������������������������������������������������������������������������������������������������� 178 References������������������������������������������������������������������������������������������������������������������� 179 16Laparoscopic One Anastomosis Gastric Bypass: History of the Procedure Surgical Technique and Outcomes��������������������������������������������������������� 181 Helmuth T. Billy, Moataz M. Bashah, and Ryan Fairley History������������������������������������������������������������������������������������������������������������������������� 181 Introduction����������������������������������������������������������������������������������������������������������������� 182 Patient Selection and Preparation������������������������������������������������������������������������������� 183 Surgical Technique����������������������������������������������������������������������������������������������������� 184 Creating the Gastric Pouch����������������������������������������������������������������������������������������� 184 Reversal of One Anastomosis Gastric Bypass ����������������������������������������������������������� 187 Benefits of a Single Anastomosis Procedure ������������������������������������������������������������� 188 Complications������������������������������������������������������������������������������������������������������������� 189 Resolution of Comorbidities and Nutritional Complications������������������������������������� 191 Outcome Comparison Roux-Y Gastric Bypass vs Sleeve Gastrectomy vs One Anastomosis Gastric Bypass��������������������������������������������������������������������������������������� 191 Insurance Coverage USA������������������������������������������������������������������������������������������� 192 Summary��������������������������������������������������������������������������������������������������������������������� 192 Question Section��������������������������������������������������������������������������������������������������������� 192 References������������������������������������������������������������������������������������������������������������������� 193 Part III Management of Bariatric Complications 17Management of Gastrointestinal Leaks and Fistula����������������������������������������������� 197 Ninh T. Nguyen and Shaun C. Daly Introduction����������������������������������������������������������������������������������������������������������������� 197 Etiologies ������������������������������������������������������������������������������������������������������������������� 197 Presentation and Diagnostic Evaluation��������������������������������������������������������������������� 198 Management��������������������������������������������������������������������������������������������������������������� 199 Nonoperative Treatment��������������������������������������������������������������������������������������������� 199 Reoperation and Drainage������������������������������������������������������������������������������������������� 199 Endoscopic Stent��������������������������������������������������������������������������������������������������������� 200 Endoscopic Stent Technique��������������������������������������������������������������������������������������� 200 Chronic Fistula����������������������������������������������������������������������������������������������������������� 202 Conclusion ����������������������������������������������������������������������������������������������������������������� 202 Question Section��������������������������������������������������������������������������������������������������������� 202 References������������������������������������������������������������������������������������������������������������������� 202 18Gastrointestinal Obstruction After Bariatric Surgery ������������������������������������������� 205 Neil A. King and Daniel M. Herron Introduction����������������������������������������������������������������������������������������������������������������� 205 Obstruction After Roux-En-Y Gastric Bypass����������������������������������������������������������� 205 Gastrojejunal Stricture ����������������������������������������������������������������������������������������������� 206 Obstruction from Internal Hernia������������������������������������������������������������������������������� 207 Small Bowel Obstruction from Scars and Adhesive Bands ��������������������������������������� 209 Incisional Hernia��������������������������������������������������������������������������������������������������������� 209 Intussusception����������������������������������������������������������������������������������������������������������� 209 Obstruction from Intraluminal Blood Clot or Bezoar������������������������������������������������� 210 xii Contents General Approach to the Bypass Patient with Obstruction����������������������������������������� 210 Obstruction in the Laparoscopic Adjustable Gastric Band Patient����������������������������� 210 Obstruction After Laparoscopic Adjustable Gastric Band Placement ����������������������� 212 Early Postoperative Band Obstruction����������������������������������������������������������������������� 212 Late Postoperative Band Obstruction������������������������������������������������������������������������� 212 Unusual Types of Band Obstruction��������������������������������������������������������������������������� 212 Obstruction After Sleeve Gastrectomy����������������������������������������������������������������������� 213 Obstruction After Biliopancreatic Diversion with Duodenal Switch������������������������� 213 Obstruction from Implanted Medical Devices����������������������������������������������������������� 214 Conclusion ����������������������������������������������������������������������������������������������������������������� 214 Question Section��������������������������������������������������������������������������������������������������������� 214 References������������������������������������������������������������������������������������������������������������������� 214 19Postoperative Bleeding in the Bariatric Surgery Patient ��������������������������������������� 217 Federico J. Serrot, Samuel Szomstein, and Raul J. Rosenthal Introduction����������������������������������������������������������������������������������������������������������������� 217 Definition/Causes������������������������������������������������������������������������������������������������������� 217 Diagnostic Algorithm������������������������������������������������������������������������������������������������� 218 Treatment Strategies��������������������������������������������������������������������������������������������������� 219 Special Considerations����������������������������������������������������������������������������������������������� 221 Conclusion ����������������������������������������������������������������������������������������������������������������� 222 Question Section��������������������������������������������������������������������������������������������������������� 222 References������������������������������������������������������������������������������������������������������������������� 222 20Management of Marginal Ulcers������������������������������������������������������������������������������� 225 Richard M. Peterson and Jason W. Kempenich Introduction����������������������������������������������������������������������������������������������������������������� 225 Pathophysiology��������������������������������������������������������������������������������������������������������� 225 Etiology����������������������������������������������������������������������������������������������������������������������� 226 Diagnosis��������������������������������������������������������������������������������������������������������������������� 227 Medical Treatment ����������������������������������������������������������������������������������������������������� 228 Prevention������������������������������������������������������������������������������������������������������������������� 228 Endoscopic and Surgical Treatment��������������������������������������������������������������������������� 229 Question Section��������������������������������������������������������������������������������������������������������� 233 References������������������������������������������������������������������������������������������������������������������� 233 21Gastric Banding Complications: Management ������������������������������������������������������� 235 Brittany Nowak, Christine Ren-Fielding, and Jeff Allen Introduction����������������������������������������������������������������������������������������������������������������� 235 Early Complications��������������������������������������������������������������������������������������������������� 235 Late Complications����������������������������������������������������������������������������������������������������� 236 Conclusion ����������������������������������������������������������������������������������������������������������������� 243 Question Section��������������������������������������������������������������������������������������������������������� 244 References������������������������������������������������������������������������������������������������������������������� 244 22Management of Nutritional Complications ������������������������������������������������������������� 247 Michael Choi, Liz Goldenberg, and Alfons Pomp Introduction����������������������������������������������������������������������������������������������������������������� 247 Macronutrients ����������������������������������������������������������������������������������������������������������� 247 Micronutrients������������������������������������������������������������������������������������������������������������� 248 Conclusions����������������������������������������������������������������������������������������������������������������� 253 Question Section��������������������������������������������������������������������������������������������������������� 255 References������������������������������������������������������������������������������������������������������������������� 255 Contents xiii 23Early and Late Dumping Syndromes����������������������������������������������������������������������� 257 Samer G. Mattar and Ann M. Rogers Introduction����������������������������������������������������������������������������������������������������������������� 257 Diagnosis��������������������������������������������������������������������������������������������������������������������� 257 Treatment ������������������������������������������������������������������������������������������������������������������� 258 Conclusions����������������������������������������������������������������������������������������������������������������� 260 Question Section��������������������������������������������������������������������������������������������������������� 260 References������������������������������������������������������������������������������������������������������������������� 260 Part IV Reoperative Bariatric Surgery for Weight Regain and Complications 24Reoperative Bariatric Surgery����������������������������������������������������������������������������������� 265 Rene Aleman, Emanuele Lo Menzo, Samuel Szomstein, and Raul J. Rosenthal Introduction����������������������������������������������������������������������������������������������������������������� 265 Reoperative Bariatric Surgery: Classification������������������������������������������������������������� 265 Preoperative Evaluation ��������������������������������������������������������������������������������������������� 269 Review of Operative Reports ������������������������������������������������������������������������������������� 269 Imaging and Functional Studies��������������������������������������������������������������������������������� 270 Dietary and Nutritional Assessment and Counseling������������������������������������������������� 270 Psychological Assessment and Counseling ��������������������������������������������������������������� 270 Preoperative Testing and Clearances�������������������������������������������������������������������������� 271 Approach��������������������������������������������������������������������������������������������������������������������� 271 Technical Aspects������������������������������������������������������������������������������������������������������� 271 LAGB-Related Reoperations ������������������������������������������������������������������������������������� 272 RYGB������������������������������������������������������������������������������������������������������������������������� 274 Loop Gastric Bypass��������������������������������������������������������������������������������������������������� 275 Biliopancreatic Diversion with Duodenal Switch (BPD-DS)������������������������������������� 276 Vertical Banded Gastroplasty (VBG)������������������������������������������������������������������������� 276 Sleeve Gastrectomy����������������������������������������������������������������������������������������������������� 277 Conclusion ����������������������������������������������������������������������������������������������������������������� 277 Question Section��������������������������������������������������������������������������������������������������������� 278 References������������������������������������������������������������������������������������������������������������������� 278 25Reoperative Options After Gastric Banding ����������������������������������������������������������� 281 Zeyad Loubnan, Manish Parikh, and Marina Kurian Introduction����������������������������������������������������������������������������������������������������������������� 281 Preoperative Workup��������������������������������������������������������������������������������������������������� 281 Surgical Options��������������������������������������������������������������������������������������������������������� 282 Outcomes ������������������������������������������������������������������������������������������������������������������� 282 Other Less Common Surgical Options for Conversion ��������������������������������������������� 284 Conclusions����������������������������������������������������������������������������������������������������������������� 285 Question Section��������������������������������������������������������������������������������������������������������� 285 References������������������������������������������������������������������������������������������������������������������� 286 26Reoperative Options After Sleeve Gastrectomy������������������������������������������������������� 287 Jacques M. Himpens, Gregg H. Jossart, and Dafydd A. Davies Introduction����������������������������������������������������������������������������������������������������������������� 287 Reasons for Inadequate Response������������������������������������������������������������������������������� 287 Rates of Failure����������������������������������������������������������������������������������������������������������� 289 Demonstration of Aberrant Anatomy and Consequences for Surgical Treatment ����� 289 Managing Inadequate Response��������������������������������������������������������������������������������� 289 xiv Contents Conclusion ����������������������������������������������������������������������������������������������������������������� 294 Question Section��������������������������������������������������������������������������������������������������������� 294 References������������������������������������������������������������������������������������������������������������������� 294 27Reoperative Options After Gastric Bypass��������������������������������������������������������������� 297 Abraham Krikhely and Marc Bessler Introduction����������������������������������������������������������������������������������������������������������������� 297 Evaluating a Patient with Weight Regain/Failure to Lose Weight ����������������������������� 297 Endoscopic Interventions������������������������������������������������������������������������������������������� 298 Surgical Revision Options ����������������������������������������������������������������������������������������� 298 Algorithms ����������������������������������������������������������������������������������������������������������������� 306 Question Section��������������������������������������������������������������������������������������������������������� 307 References������������������������������������������������������������������������������������������������������������������� 307 28Revisional Bariatric Surgery for Management of Late Complications����������������� 309 Patrick J. Sweigert, Fadi Bakhos, Eric Marcotte, and Bipan Chand Introduction����������������������������������������������������������������������������������������������������������������� 309 Late Complications Following Sleeve Gastrectomy Requiring Revision������������������� 309 Late Complications Following Roux-en-Y Gastric Bypass Requiring Revision������� 313 Late Complications Following Biliopancreatic Diversion with Duodenal Switch Requiring Revision����������������������������������������������������������������������������������������������������� 317 Conclusion ����������������������������������������������������������������������������������������������������������������� 318 Question Section��������������������������������������������������������������������������������������������������������� 318 References������������������������������������������������������������������������������������������������������������������� 318 29Revisional Surgery Data and Guidelines ����������������������������������������������������������������� 321 Kunoor Jain-Spangler and Ranjan Sudan Introduction����������������������������������������������������������������������������������������������������������������� 321 Inadequate Weight Loss ��������������������������������������������������������������������������������������������� 322 Weight Regain������������������������������������������������������������������������������������������������������������� 323 Lack of Comorbidity Resolution or Return of Comorbidities ����������������������������������� 323 Anatomic Complications ������������������������������������������������������������������������������������������� 323 Approach to the Reoperative Surgery Patient������������������������������������������������������������� 324 Summary��������������������������������������������������������������������������������������������������������������������� 324 Question Section��������������������������������������������������������������������������������������������������������� 325 References������������������������������������������������������������������������������������������������������������������� 325 Part V Metabolic Surgery 30Operation of Choice for Metabolic Surgery������������������������������������������������������������� 329 Ali Aminian and Philip R. Schauer Introduction����������������������������������������������������������������������������������������������������������������� 329 Standard Metabolic Surgical Procedures ������������������������������������������������������������������� 332 Clinical Outcomes������������������������������������������������������������������������������������������������������� 332 Mechanism of Action������������������������������������������������������������������������������������������������� 332 Comparison of Metabolic Procedures������������������������������������������������������������������������� 335 RCTs Comparing RYGB to SG ��������������������������������������������������������������������������������� 335 Meta-analysis of RCTs Comparing RYGB to SG for T2DM������������������������������������� 336 Individualized Metabolic Surgery Score��������������������������������������������������������������������� 337 Other Considerations in Decision-Making Between RYGB and SG������������������������� 338 Summary��������������������������������������������������������������������������������������������������������������������� 339 Question Section��������������������������������������������������������������������������������������������������������� 339 References������������������������������������������������������������������������������������������������������������������� 340 Contents xv 31Outcomes of Metabolic Surgery ������������������������������������������������������������������������������� 341 Rene Aleman, Francesco Rubino, Emanuele Lo Menzo, and Raul J. Rosenthal Introduction: Definition of Metabolic Surgery����������������������������������������������������������� 341 Outcomes of Metabolic Surgery: Retrospective and Observational Studies ������������� 342 Outcomes of Metabolic Surgery: Randomized Controlled Trials������������������������������� 347 Conclusions����������������������������������������������������������������������������������������������������������������� 349 Question Section��������������������������������������������������������������������������������������������������������� 349 References������������������������������������������������������������������������������������������������������������������� 349 32Operative Outcomes of Metabolic/Bariatric Surgery in Subjects with Type 1 Obesity Index (30–35 kg/m2) ������������������������������������������������������������������������������������� 353 Ricardo V. Cohen, Tarissa Z. Petry, and Estefano A. Negri Introduction����������������������������������������������������������������������������������������������������������������� 353 Outcomes ������������������������������������������������������������������������������������������������������������������� 353 Innovative Procedures������������������������������������������������������������������������������������������������� 356 Conclusions����������������������������������������������������������������������������������������������������������������� 356 Question Section��������������������������������������������������������������������������������������������������������� 356 References������������������������������������������������������������������������������������������������������������������� 357 Part VI Endoscopic Bariatric Surgery 33The Role of Preoperative Endoscopy in Bariatric Surgery������������������������������������� 361 Daniel Davila Bradley and Kevin M. Reavis Introduction����������������������������������������������������������������������������������������������������������������� 361 General Considerations����������������������������������������������������������������������������������������������� 362 Preprocedure Setup and Upper Endoscopy Technique����������������������������������������������� 363 Preoperative Endoscopy in Patients Undergoing Index Procedures��������������������������� 363 Preoperative Endoscopy in Revisional Patients ��������������������������������������������������������� 364 Preoperative Endoscopy in Revisional Patients: Marginal Ulcer������������������������������� 364 Preoperative Endoscopy in Revisional Patients: Stricture ����������������������������������������� 365 Preoperative Endoscopy in Revisional Patients: Postoperative Leaks����������������������� 365 Preoperative Endoscopy in Revisional Patients: Band Erosion, Impaction, and Slippage��������������������������������������������������������������������������������������������� 366 Preoperative Endoscopy in Revisional Patients: Gastroesophageal Reflux Disease After Bypass or Biliopancreatic Diversion with Duodenal Switch��������������� 366 Preoperative Endoscopy in Revisional Patients: Previous Endoscopic Procedures��� 367 Preoperative Endoscopy in Revisional Patients: Biliopancreatic Access������������������� 368 Forensic Endoscopy ��������������������������������������������������������������������������������������������������� 369 Forensic Endoscopy: Malabsorptive��������������������������������������������������������������������������� 369 Forensic Endoscopy: Restrictive and Malabsorptive ������������������������������������������������� 370 Forensic Endoscopy: Restrictive��������������������������������������������������������������������������������� 371 Collaborative Efforts��������������������������������������������������������������������������������������������������� 372 Conclusion ����������������������������������������������������������������������������������������������������������������� 373 Question Section��������������������������������������������������������������������������������������������������������� 373 References������������������������������������������������������������������������������������������������������������������� 373 34Intragastric Balloon Therapy ����������������������������������������������������������������������������������� 375 Jaime Ponce and Rami E. Lutfi Background and History��������������������������������������������������������������������������������������������� 375 FDA-Approved Balloons ������������������������������������������������������������������������������������������� 376 Placement and Removal Technique ��������������������������������������������������������������������������� 376 Patient Management��������������������������������������������������������������������������������������������������� 379 Outcomes and Complications������������������������������������������������������������������������������������� 379 Future Technologies��������������������������������������������������������������������������������������������������� 381 xvi Contents Conclusions����������������������������������������������������������������������������������������������������������������� 381 Question Section��������������������������������������������������������������������������������������������������������� 381 References������������������������������������������������������������������������������������������������������������������� 381 35Endoluminal Gastric Pouch Revision����������������������������������������������������������������������� 383 Brian Hodgens, Simon Che, and Dean J. Mikami Introduction����������������������������������������������������������������������������������������������������������������� 383 Endoscopic Treatment������������������������������������������������������������������������������������������������� 384 Conclusion ����������������������������������������������������������������������������������������������������������������� 388 Question Section��������������������������������������������������������������������������������������������������������� 388 References������������������������������������������������������������������������������������������������������������������� 388 36Endoscopic Primary Bariatric Procedures��������������������������������������������������������������� 391 Michelle H. Scerbo, Melissa M. Felinski, Kulvinder S. Bajwa, Erik B. Wilson, and Shinil K. Shah Introduction����������������������������������������������������������������������������������������������������������������� 391 Gastric Procedures ����������������������������������������������������������������������������������������������������� 392 Gastric Balloons��������������������������������������������������������������������������������������������������������� 392 Gastroplasty ��������������������������������������������������������������������������������������������������������������� 392 Other Gastric Procedures ������������������������������������������������������������������������������������������� 396 Small Bowel Procedures��������������������������������������������������������������������������������������������� 398 Duodenal Mucosal Surfacing������������������������������������������������������������������������������������� 400 Conclusion ����������������������������������������������������������������������������������������������������������������� 400 Question Section��������������������������������������������������������������������������������������������������������� 401 References������������������������������������������������������������������������������������������������������������������� 401 37Endoscopic Management of Stomal Stenosis����������������������������������������������������������� 403 Crystal E. Alvarez and Keith Scharf Introduction����������������������������������������������������������������������������������������������������������������� 403 History of Endoscopy������������������������������������������������������������������������������������������������� 403 Preprocedure Room Setup and Endoscopic Technique ��������������������������������������������� 404 Sleeve Gastrectomy����������������������������������������������������������������������������������������������������� 405 Treatment of Sleeve Stenosis ������������������������������������������������������������������������������������� 406 Roux-en-Y Gastric Bypass����������������������������������������������������������������������������������������� 407 Treatment Options for RYGB Stenosis����������������������������������������������������������������������� 409 Conclusion ����������������������������������������������������������������������������������������������������������������� 411 Question Section��������������������������������������������������������������������������������������������������������� 411 References������������������������������������������������������������������������������������������������������������������� 411 Part VII Quality in Bariatric Surgery 38Patient Safety��������������������������������������������������������������������������������������������������������������� 417 Mohamad Rassoul A. Abu-Nuwar, Robert B. Lim, and Daniel B. Jones Introduction����������������������������������������������������������������������������������������������������������������� 417 Defining Safety����������������������������������������������������������������������������������������������������������� 417 Ensuring Quality and Improvement��������������������������������������������������������������������������� 419 Preoperative Assessment��������������������������������������������������������������������������������������������� 420 Perioperative Care������������������������������������������������������������������������������������������������������� 421 Postoperative Care ����������������������������������������������������������������������������������������������������� 424 Clinical Pathways and Teamwork������������������������������������������������������������������������������� 424 Disclosure of Complications and Error in Post-Adverse Event Management����������� 427 Conclusion ����������������������������������������������������������������������������������������������������������������� 427 Question Section��������������������������������������������������������������������������������������������������������� 428 References������������������������������������������������������������������������������������������������������������������� 428 Contents xvii 39LABS Project��������������������������������������������������������������������������������������������������������������� 431 Bruce M. Wolfe and Elizaveta Walker Introduction to the Problem ��������������������������������������������������������������������������������������� 431 History of the LABS Consortium������������������������������������������������������������������������������� 431 Goals of the LABS Consortium��������������������������������������������������������������������������������� 432 Rationale for Research Design����������������������������������������������������������������������������������� 433 Structure and Working Organization of LABS����������������������������������������������������������� 433 Outcome Domains in Bariatric Surgery��������������������������������������������������������������������� 434 Weight Loss and Body Composition ������������������������������������������������������������������������� 434 Diabetes Mellitus and Insulin Resistance������������������������������������������������������������������� 436 Cardiovascular and Pulmonary Disease��������������������������������������������������������������������� 436 Renal Disease������������������������������������������������������������������������������������������������������������� 436 Liver Function������������������������������������������������������������������������������������������������������������� 436 Behavioral/Psychosocial Factors ������������������������������������������������������������������������������� 436 Musculoskeletal and Functional Status����������������������������������������������������������������������� 437 Gender Issues ������������������������������������������������������������������������������������������������������������� 437 Economic Impact ������������������������������������������������������������������������������������������������������� 437 Biospecimens ������������������������������������������������������������������������������������������������������������� 437 Results from the LABS Consortium��������������������������������������������������������������������������� 437 LABS-1 Baseline Data����������������������������������������������������������������������������������������������� 437 LABS-1 Results ��������������������������������������������������������������������������������������������������������� 438 LABS-1 and LABS-2: 30-Day Results����������������������������������������������������������������������� 439 LABS-2 Baseline Data Reports ��������������������������������������������������������������������������������� 440 LABS-2 Outcome Studies������������������������������������������������������������������������������������������� 442 Conclusion ����������������������������������������������������������������������������������������������������������������� 444 Question Section��������������������������������������������������������������������������������������������������������� 445 References������������������������������������������������������������������������������������������������������������������� 446 40Quality in Bariatric Surgery ������������������������������������������������������������������������������������� 449 Robin P. Blackstone, Thomas P. Petrick, and Anthony T. Petrick Introduction����������������������������������������������������������������������������������������������������������������� 449 The History of Quality in Bariatric Surgery��������������������������������������������������������������� 449 American Society for Metabolic and Bariatric Surgery Bariatric Surgery Center of Excellence (ASMBS BSCOE) Program����������������������������������������������������� 450 American College of Surgeons Bariatric Surgery Center Network (ACS BSCN)����� 451 The Michagan Bariatric Surgical Collaborative (MiBSC)����������������������������������������� 451 The Evolution of the ASMBS BSCOE����������������������������������������������������������������������� 452 The Maturation of MBSAQIP������������������������������������������������������������������������������������� 454 Additional ASMBS Quality Initiatives����������������������������������������������������������������������� 458 The MBSAQIP Approach to Accreditation����������������������������������������������������������������� 458 The Future of Quality in Bariatric Surgery����������������������������������������������������������������� 469 Question Section��������������������������������������������������������������������������������������������������������� 470 References������������������������������������������������������������������������������������������������������������������� 470 41Patient Experience and Perioperative Pathway in Bariatric Surgery������������������� 473 Nabeel R. Obeid, Ryan Howard, and Dana A. Telem Introduction����������������������������������������������������������������������������������������������������������������� 473 Access to Care������������������������������������������������������������������������������������������������������������� 473 Program Pathways and Patient Experience����������������������������������������������������������������� 474 Perioperative Care������������������������������������������������������������������������������������������������������� 475 Postoperative Care ����������������������������������������������������������������������������������������������������� 478 Question Section��������������������������������������������������������������������������������������������������������� 481 References������������������������������������������������������������������������������������������������������������������� 482 xviii 42Decreasing Readmissions in Bariatric Surgery������������������������������������������������������� 487 John M. Morton Introduction����������������������������������������������������������������������������������������������������������������� 487 Rationale��������������������������������������������������������������������������������������������������������������������� 487 Mechanisms for Decreasing Readmission Rates ������������������������������������������������������� 487 Team Approach����������������������������������������������������������������������������������������������������������� 488 Characterizing Readmissions Following Bariatric Surgery��������������������������������������� 489 Discussion������������������������������������������������������������������������������������������������������������������� 491 Next Steps������������������������������������������������������������������������������������������������������������������� 492 Question Section��������������������������������������������������������������������������������������������������������� 493 References������������������������������������������������������������������������������������������������������������������� 493 Part VIII Specific Considerations 43Enhanced Recovery in Bariatric Surgery����������������������������������������������������������������� 497 Xiaoxi (Chelsea) Feng and Stacy A. Brethauer Introduction����������������������������������������������������������������������������������������������������������������� 497 Pathophysiological Principles������������������������������������������������������������������������������������� 497 Practice Implementation��������������������������������������������������������������������������������������������� 498 Enhanced Recovery Outcomes in Bariatric Surgery��������������������������������������������������� 499 Exclusion and Barriers to Implementation����������������������������������������������������������������� 500 Economic Benefits ����������������������������������������������������������������������������������������������������� 502 Conclusion ����������������������������������������������������������������������������������������������������������������� 502 Question Section��������������������������������������������������������������������������������������������������������� 502 References������������������������������������������������������������������������������������������������������������������� 503 44Biliary Tract Disease in the Bariatric Surgery Patient������������������������������������������� 505 Adam C. Sheka, Keith M. Wirth, and Sayeed Ikramuddin Introduction����������������������������������������������������������������������������������������������������������������� 505 Pathophysiology of Cholesterol Cholelithiasis����������������������������������������������������������� 505 Epidemiology of Cholelithiasis���������������������������������������������������������������������������������� 506 Rapid Weight Loss and Gallstone Formation������������������������������������������������������������� 507 Incidence of Cholelithiasis in Bariatric Surgery Patients������������������������������������������� 507 Challenges in Management of the Gallbladder at the Time of Bariatric Operation��� 507 Management Pathways in Patients Undergoing Bariatric Surgery����������������������������� 508 Pharmacologic Prevention of Cholelithiasis After Bariatric Surgery������������������������� 508 Access to the Biliary Tree Following Roux-­en-­Y Gastric Bypass����������������������������� 509 Conclusions����������������������������������������������������������������������������������������������������������������� 511 Question Section��������������������������������������������������������������������������������������������������������� 511 References������������������������������������������������������������������������������������������������������������������� 511 45Joint Disease and Obesity: Opportunity for Multidisciplinary Investigation and Collaboration ������������������������������������������������������������������������������� 515 John M. Morton Rationale��������������������������������������������������������������������������������������������������������������������� 515 Risk of Obesity for Joint Disease and Joint Replacement Surgery���������������������������� 515 Bariatric and Orthopedic Surgery: Team Approach��������������������������������������������������� 516 Question Section��������������������������������������������������������������������������������������������������������� 517 References������������������������������������������������������������������������������������������������������������������� 517 46Cardiac Risk Factor Improvement Following Bariatric Surgery��������������������������� 519 Riley Katsuki Kitamura, John M. Morton, and Dan Eisenberg Introduction����������������������������������������������������������������������������������������������������������������� 519 Obesity and Cardiovascular Disease��������������������������������������������������������������������������� 520 Contents Contents xix Preoperative Cardiac Evaluation��������������������������������������������������������������������������������� 522 Bariatric Surgery and Cardiac Risk Factors��������������������������������������������������������������� 522 Conclusion ����������������������������������������������������������������������������������������������������������������� 523 Question Section��������������������������������������������������������������������������������������������������������� 523 References������������������������������������������������������������������������������������������������������������������� 524 47Critical Care Considerations in the Bariatric Patient��������������������������������������������� 527 Stacy A. Brethauer, Lucia H. Nguyen, and David A. Provost Introduction����������������������������������������������������������������������������������������������������������������� 527 Neurologic������������������������������������������������������������������������������������������������������������������� 527 Pulmonary������������������������������������������������������������������������������������������������������������������� 528 Cardiovascular ����������������������������������������������������������������������������������������������������������� 529 Gastrointestinal����������������������������������������������������������������������������������������������������������� 529 Nutrition��������������������������������������������������������������������������������������������������������������������� 529 Renal��������������������������������������������������������������������������������������������������������������������������� 530 Endocrine ������������������������������������������������������������������������������������������������������������������� 530 Infectious Disease������������������������������������������������������������������������������������������������������� 530 Pharmacology������������������������������������������������������������������������������������������������������������� 531 Hematology����������������������������������������������������������������������������������������������������������������� 531 Conclusions����������������������������������������������������������������������������������������������������������������� 532 Question Section��������������������������������������������������������������������������������������������������������� 532 References������������������������������������������������������������������������������������������������������������������� 532 48Bariatric Surgery in Adolescents������������������������������������������������������������������������������� 535 S. Christopher Derderian, Marc P. Michalsky, and Thomas H. Inge Introduction����������������������������������������������������������������������������������������������������������������� 535 Definition of Pediatric Obesity����������������������������������������������������������������������������������� 535 Consequences of Obesity in Adolescence������������������������������������������������������������������� 535 Cardiovascular Disease����������������������������������������������������������������������������������������������� 536 Nonalcoholic Fatty Liver Disease������������������������������������������������������������������������������� 536 Glucose Impairment��������������������������������������������������������������������������������������������������� 537 Obstructive Sleep Apnea��������������������������������������������������������������������������������������������� 537 Idiopathic Intracranial Hypertension (IIH)����������������������������������������������������������������� 537 Psychological and Quality of Life Issues������������������������������������������������������������������� 537 Musculoskeletal Disorders����������������������������������������������������������������������������������������� 537 Pharmacotherapy��������������������������������������������������������������������������������������������������������� 537 Incidence and Best Practice Guidelines for Adolescent MBS ����������������������������������� 538 Outcomes of Metabolic and Bariatric Surgery in Adolescents����������������������������������� 540 Summary��������������������������������������������������������������������������������������������������������������������� 541 Question Section��������������������������������������������������������������������������������������������������������� 541 References������������������������������������������������������������������������������������������������������������������� 542 49Pregnancy Issues and Bariatric Surgery ����������������������������������������������������������������� 545 Tripurari Mishra and Shanu N. Kothari Introduction����������������������������������������������������������������������������������������������������������������� 545 Effects of Prepregnancy Obesity on Fertility and Pregnancy Outcomes ������������������� 545 Obesity and Polycystic Ovarian Syndrome ��������������������������������������������������������������� 546 Impact of Bariatric Surgery on Fertility��������������������������������������������������������������������� 547 Impact of Bariatric Surgery on Mother and Neonate������������������������������������������������� 547 Nutrition Factors After Bariatric Surgery������������������������������������������������������������������� 548 Internal Hernia During Pregnancy ����������������������������������������������������������������������������� 548 Conclusion ����������������������������������������������������������������������������������������������������������������� 551 Question Section��������������������������������������������������������������������������������������������������������� 551 References������������������������������������������������������������������������������������������������������������������� 552 xx 50Robotics in Bariatric Surgery ����������������������������������������������������������������������������������� 553 Keith Chae Kim, Jonathan Douissard, Cynthia K. Buffington, and Monika E. Hagen Introduction����������������������������������������������������������������������������������������������������������������� 553 Robotics for Bariatric Surgery ����������������������������������������������������������������������������������� 553 Robotic Roux-en-Y Gastric Bypass (RYGB)������������������������������������������������������������� 554 Robotic Sleeve Gastrectomy (SG) ����������������������������������������������������������������������������� 554 Robotic Adjustable Gastric Band (AGB)������������������������������������������������������������������� 555 Robotic Biliopancreatic Diversion with Duodenal Switch (BPD/DS)����������������������� 555 Robotic Revisional Surgery ��������������������������������������������������������������������������������������� 556 Cost of Robotic Bariatric Surgery������������������������������������������������������������������������������� 556 Currently Available Robotic Surgery Systems����������������������������������������������������������� 557 Emerging Robotic Surgery Systems��������������������������������������������������������������������������� 558 Conclusion ����������������������������������������������������������������������������������������������������������������� 559 Question Section��������������������������������������������������������������������������������������������������������� 559 References������������������������������������������������������������������������������������������������������������������� 560 51Body Contouring After Massive Weight Loss ��������������������������������������������������������� 563 Natalie S. Barton, Al S. Aly, and Gregory R. D. Evans Introduction����������������������������������������������������������������������������������������������������������������� 563 Presentation of the Massive Weight Loss Patient to the Plastic Surgeon������������������� 563 Workup for Post-Massive Weight Loss Plastic Surgery��������������������������������������������� 564 Postoperative Management����������������������������������������������������������������������������������������� 566 The Lower Trunk ������������������������������������������������������������������������������������������������������� 566 The Upper Arms��������������������������������������������������������������������������������������������������������� 567 The Upper Trunk��������������������������������������������������������������������������������������������������������� 570 Thighs������������������������������������������������������������������������������������������������������������������������� 571 Miscellaneous Regions����������������������������������������������������������������������������������������������� 573 Important Issues for the Bariatric Surgeon/Plastic Surgeon Interaction��������������������� 573 Conclusion ����������������������������������������������������������������������������������������������������������������� 574 Question Section��������������������������������������������������������������������������������������������������������� 574 References������������������������������������������������������������������������������������������������������������������� 575 52The Practice of Bariatric Coding and Reimbursement������������������������������������������� 577 Laura Dewender and Ashutosh Kaul Introduction����������������������������������������������������������������������������������������������������������������� 577 From Global Proportions, Trend, and Cost Issues to Surgical Intervention��������������� 577 The Value of Surgery: The Basics of RVUs and CPTs����������������������������������������������� 578 Bariatric Surgery CPT Codes������������������������������������������������������������������������������������� 579 Coding Nuances ��������������������������������������������������������������������������������������������������������� 579 Insurance Preauthorization: Initial Bariatric Surgery������������������������������������������������� 580 Post-surgery Appeals�������������������������������������������������������������������������������������������������� 580 Participation in Specific Plans ����������������������������������������������������������������������������������� 582 Contracted Rates��������������������������������������������������������������������������������������������������������� 582 Conclusion ����������������������������������������������������������������������������������������������������������������� 582 Question Section��������������������������������������������������������������������������������������������������������� 582 References������������������������������������������������������������������������������������������������������������������� 583 53Medical Malpractice in Bariatric Surgery: The ASMBS Journey to a Closed Claims Registry����������������������������������������������������������������������������������������������������������� 585 William A. Sweet and Eric J. DeMaria The Challenge Defined����������������������������������������������������������������������������������������������� 585 ASMBS Efforts����������������������������������������������������������������������������������������������������������� 585 Most Important “Quality Care” Deficiencies and Outcomes Prompting Bariatric Claims ��������������������������������������������������������������������������������������������������������� 587 Contents Contents xxi The Critical Importance of “History”������������������������������������������������������������������������� 587 Excellence in Consent Process, Rapport-­Building: The Communication Keys Prior to “Not Guilty” After an Adverse Event��������������������������������������������������� 587 “Anatomy of a Near Miss,” the Sometimes Critical Difficulty of In-Hospital Communication����������������������������������������������������������������������������������������������������������� 588 Disastrous Team Training and Handoff Management: Don’t Fumble the Handoff��� 589 The Substantial Hazards of Inadequate Postoperative Communications Planning ��������������������������������������������������������������������������������������������������������������������� 589 GI Bleed Post-RYGB Management Disaster, the most read�������������������������������������� 590 Delay/Failure to Treat and Communication Lapses Are Frequently Paired��������������� 592 A Leak Event Compounded��������������������������������������������������������������������������������������� 592 Question Section��������������������������������������������������������������������������������������������������������� 593 References������������������������������������������������������������������������������������������������������������������� 593 54Obesity Prevention����������������������������������������������������������������������������������������������������� 595 Elizaveta Walker and Bruce M. Wolfe Introduction����������������������������������������������������������������������������������������������������������������� 595 Obesity and Its Causes ����������������������������������������������������������������������������������������������� 595 Prevention Strategies in Adult Populations����������������������������������������������������������������� 597 Prevention Strategies in Childhood and Adolescent Populations������������������������������� 601 Monitoring and Surveillance of Obesity��������������������������������������������������������������������� 604 Resources ������������������������������������������������������������������������������������������������������������������� 606 Conclusion ����������������������������������������������������������������������������������������������������������������� 607 Question Section��������������������������������������������������������������������������������������������������������� 607 References������������������������������������������������������������������������������������������������������������������� 607 55Training in Bariatric Surgery ����������������������������������������������������������������������������������� 613 Corrigan L. McBride Background����������������������������������������������������������������������������������������������������������������� 613 Evolution of Bariatric Surgery Fellowship Training��������������������������������������������������� 614 Question Section��������������������������������������������������������������������������������������������������������� 616 References������������������������������������������������������������������������������������������������������������������� 617 56Adjuvant Pharmaceutical Therapy for Perioperative Use in Bariatric Surgery ������������������������������������������������������������������������������������������������������� 619 John M. Morton, Saber Ghiassi, and Geoffrey S. Nadzam Introduction����������������������������������������������������������������������������������������������������������������� 619 Rationale: Variation in Treatment Effect��������������������������������������������������������������������� 619 Preoperative Weight Loss������������������������������������������������������������������������������������������� 620 Postoperative Weight Gain����������������������������������������������������������������������������������������� 621 Anti-obesity Medications: Indications and Contraindications����������������������������������� 621 Adjuvant Pharmaceutical Therapy with Bariatric Surgery����������������������������������������� 622 Next Steps������������������������������������������������������������������������������������������������������������������� 622 Conclusion ����������������������������������������������������������������������������������������������������������������� 623 Question Section��������������������������������������������������������������������������������������������������������� 623 References������������������������������������������������������������������������������������������������������������������� 623 Answers������������������������������������������������������������������������������������������������������������������������������� 625 Answers (Chap. 1) ������������������������������������������������������������������������������������������������������� 625 Answers (Chap. 2) ������������������������������������������������������������������������������������������������������� 625 Answers (Chap. 5) ������������������������������������������������������������������������������������������������������� 626 Answers (Chap. 6) ������������������������������������������������������������������������������������������������������� 626 Answers (Chap. 7) ������������������������������������������������������������������������������������������������������� 626 Answers (Chap. 8) ������������������������������������������������������������������������������������������������������� 626 Answers (Chap. 9) ������������������������������������������������������������������������������������������������������� 627 xxii Answers (Chap. 10) ����������������������������������������������������������������������������������������������������� 627 Answers (Chap. 11) ����������������������������������������������������������������������������������������������������� 627 Answers (Chap. 12) ����������������������������������������������������������������������������������������������������� 628 Answers (Chap. 13) ����������������������������������������������������������������������������������������������������� 628 Answers (Chap. 14) ����������������������������������������������������������������������������������������������������� 628 Answers (Chap. 15) ����������������������������������������������������������������������������������������������������� 628 Answers (Chap. 16) ����������������������������������������������������������������������������������������������������� 629 Answers (Chap. 17) ����������������������������������������������������������������������������������������������������� 629 Answers (Chap. 18) ����������������������������������������������������������������������������������������������������� 629 Answers (Chap. 19) ����������������������������������������������������������������������������������������������������� 629 Answers (Chap. 20) ����������������������������������������������������������������������������������������������������� 629 Answers (Chap. 21) ����������������������������������������������������������������������������������������������������� 630 Answers (Chap. 22) ����������������������������������������������������������������������������������������������������� 630 Answers (Chap. 23) ����������������������������������������������������������������������������������������������������� 630 Answers (Chap. 24) ����������������������������������������������������������������������������������������������������� 630 Answers (Chap. 25) ����������������������������������������������������������������������������������������������������� 631 Answers (Chap. 26) ����������������������������������������������������������������������������������������������������� 631 Answers (Chap. 27) ����������������������������������������������������������������������������������������������������� 631 Answers (Chap. 28) ����������������������������������������������������������������������������������������������������� 631 Answers (Chap. 29) ����������������������������������������������������������������������������������������������������� 632 Answers (Chap. 30) ����������������������������������������������������������������������������������������������������� 632 Answers (Chap. 31) ����������������������������������������������������������������������������������������������������� 632 Answers (Chap. 32) ����������������������������������������������������������������������������������������������������� 633 Answers (Chap. 33) ����������������������������������������������������������������������������������������������������� 633 Answers (Chap. 34) ����������������������������������������������������������������������������������������������������� 633 Answers (Chap. 35) ����������������������������������������������������������������������������������������������������� 634 Answers (Chap. 36) ����������������������������������������������������������������������������������������������������� 634 Answers (Chap. 37) ����������������������������������������������������������������������������������������������������� 634 Answers (Chap. 38) ����������������������������������������������������������������������������������������������������� 634 Answers (Chap. 39) ����������������������������������������������������������������������������������������������������� 635 Answers (Chap. 40) ����������������������������������������������������������������������������������������������������� 635 Answers (Chap. 41) ����������������������������������������������������������������������������������������������������� 635 Answers (Chap. 42) ����������������������������������������������������������������������������������������������������� 635 Answers (Chap. 43) ����������������������������������������������������������������������������������������������������� 636 Answers (Chap. 44) ����������������������������������������������������������������������������������������������������� 636 Answers (Chap. 45) ����������������������������������������������������������������������������������������������������� 636 Answers (Chap. 46) ����������������������������������������������������������������������������������������������������� 636 Answers (Chap. 47) ����������������������������������������������������������������������������������������������������� 636 Answers (Chap. 48) ����������������������������������������������������������������������������������������������������� 637 Answers (Chap. 49) ����������������������������������������������������������������������������������������������������� 637 Answers (Chap. 50) ����������������������������������������������������������������������������������������������������� 637 Answers (Chap. 51) ����������������������������������������������������������������������������������������������������� 637 Answers (Chap. 52) ����������������������������������������������������������������������������������������������������� 637 Answers (Chap. 53) ����������������������������������������������������������������������������������������������������� 638 Answers (Chap. 54) ����������������������������������������������������������������������������������������������������� 638 Answers (Chap. 55) ����������������������������������������������������������������������������������������������������� 638 Answers (Chap. 56) ����������������������������������������������������������������������������������������������������� 638 Index������������������������������������������������������������������������������������������������������������������������������������� 639 Contents Contributors Mohamad Rassoul A. Abu-Nuwar, MD Division of Bariatric & Minimally Invasive Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA Rene Aleman, MD Minimally Invasive Surgery and Bariatric Surgery Department, Cleveland Clinic Florida, Weston, FL, USA Jeff Allen, MD, FACS, FASMBS General Surgery, Division of Bariatric Surgery, Norton Healthcare, Louisville, KY, USA Crystal E. Alvarez, DO Minimally Invasive Surgery, Bariatric Surgery, Department of General Surgery, Loma Linda University Health, Loma Linda, CA, USA Al S. Aly, MD Plastic Surgery, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates Ali Aminian, MD, FACS, FASMBS Department of General Surgery, Bariatric and Metabolic Institute, Cleveland Clinic, Cleveland, OH, USA Kulvinder S. Bajwa, MD Department of Surgery, Division of Minimally Invasive and Elective General Surgery, McGovern Medical School, UT Health, Houston, TX, USA Fadi Bakhos, MD Department of Surgery, Loyola University Medical Center, Maywood, IL, USA Natalie S. Barton, MD Plastic Surgery, University of California, Irvine, Orange, CA, USA Moataz M. Bashah, MD Division of Metabolic and Bariatric Surgery, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar Marc Bessler, MD Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA Monica M. Betancourt-Garcia, MD DHR Health Institute for Research and Development, DHR Health System, Edinburg, TX, USA Helmuth T. Billy, MD Metabolic and Bariatric Surgery, Community Memorial Hospital, Ventura, St. John’s Regional Medical Center, Oxnard, CA, USA Robin P. Blackstone, MD, FACS, FASMBS Metabolic and Bariatric Surgery, Banner University Medical Center – Phoenix, Phoenix, AZ, USA Aaron Bolduc, MD Department of Surgery, Augusta University Medical Center, Augusta, GA, USA Daniel Davila Bradley, MD Gastrointestinal Minimal Invasive Surgery, The Oregon Clinic, Portland, OR, USA Bariatric Surgery, Portland Providence Medical Center/Legacy Medical Center, Portland, OR, USA Stacy A. Brethauer, MD, FASMBS Department of Surgery, The Ohio State University, Columbus, OH, USA xxiii xxiv Cynthia K. Buffington, PhD Department of General Surgery/Bariatrics, Center for Metabolic and Obesity Surgery at Advent Health Celebration, Celebration, FL, USA Bipan Chand, MD, FACS, FASGE, FASMBS Department of Surgery, Division of GI/ Minimally Invasive Surgery, Loyola University Medical Center, Stritch School of Medicine, Maywood, IL, USA Simon Che, MD Department of Surgery, University of Hawaii John A. Burns School of Medicine, Honolulu, HI, USA Michael Choi, MD Department of Surgery, Weill Cornell Medical Center, New York-­ Presbyterian Hospital, New York, NY, USA Elias Chousleb, MD, FACS, FASMBS Department of Surgery, Florida International University, Herbert Wertheim College of Medicine, Miami, FL, USA Ricardo V. Cohen, MD, FASMBS, FACS The Center for Obesity and Diabetes, Oswaldo Cruz German Hospital, São Paulo, Brazil Daniel Cottam, MD Bariatric Medicine Institute, Salt Lake City, UT, USA Samuel Cottam, HS Bariatric Medicine Institute, Salt Lake City, UT, USA Shaun C. Daly, MD Department of Surgery, Division of Gastrointestinal Surgery, University of California, Irvine Medical Center, Orange, CA, USA Dafydd A. Davies, MD, MPhil, FRCSC Division of Pediatric Thoracic and General Surgery, The Hospital for Sick Children, Toronto, ON, Canada Eric J. DeMaria, MD, FACS, FASMBS American Society for Metabolic and Bariatric Surgery, Division of General/Bariatric Surgery, Department of Surgery, Brody School of Medicine, East Carolina University, Greenville, NC, USA S. Christopher Derderian, MD Pediatric Surgery, Children’s Hospital Colorado, Aurora, CO, USA Laura Dewender, CPC Coding and Compliance, Advanced Surgeons, Valhalla, NY, USA Jonathan Douissard, MD Division of Digestive Surgery, University Hospital Geneva, Geneva, Switzerland Christopher DuCoin, MD, MPH, FACS, FASMBS Division of Bariatric & General Surgery, University of South Florida, Tampa, FL, USA Dan Eisenberg, MD, MS, FASMBS Department of Surgery, Stanford School of Medicine and VA Palo Alto Health Care System, Palo Alto, CA, USA Wayne J. English, MD Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA Gregory R. D. Evans, MD, FACS Plastic Surgery and Biomedical Engineering, University of California, Irvine, Orange, CA, USA Ryan Fairley, DO Surgical Education, Community Memorial Hospital, Ventura, CA, USA Melissa M. Felinski, DO Department of Surgery, Division of Minimally Invasive and Elective General Surgery, McGovern Medical School, UT Health, Houston, TX, USA Xiaoxi (Chelsea) Feng, MD, MPH Department of General Surgery, Cleveland Clinic, Cleveland, OH, USA R. Armour Forse, MD, PhD DHR Health Institute for Education and Innovation, DHR Health System, Department of Surgery, University of Texas Rio Grande Valley, Edinburg, TX, USA Contributors Contributors xxv David Romero Funes, MD Department of General Surgery, The Bariatric and Metabolic Institute, Cleveland Clinic Florida, Weston, FL, USA Michel Gagner, MD, FRCSC, FACS Department of Surgery, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA Department of Surgery, Hôpital du Sacré-Coeur, Montreal, QC, Canada Saber Ghiassi, MD, MPH Department of Surgery, Yale School of Medicine, Fairfield, CT, USA Liz Goldenberg, MPH, RD, CDN GI Metabolic and Bariatric Surgery, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY, USA Monika E. Hagen, MD, MBA Division of Digestive Surgery, University Hospital Geneva, Geneva, Switzerland Juan D. Hernandez R., MD, FACS Surgery and Anatomy, Hospital Universitario Fundación Santa Fe de Bogotá, Universidad de los Andes School of Medicine, Bogotá, Colombia Daniel M. Herron, MD, FACS, FASMBS Garlock Division of Surgery, Mount Sinai Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA Kelvin D. Higa, MD Department of Surgery, Minimally Invasive and Bariatric Surgery, Fresno Heart and Surgical Hospital, University of California San Francisco, Fresno, CA, USA Jacques M. Himpens, MD, PhD, FASMBS (Hon) Abdominal Surgery, St. Pierre University Hospital, Brussels, Belgium Brian Hodgens, MD Department of Surgery, University of Hawaii John A. Burns School of Medicine, Honolulu, HI, USA Ryan Howard, MD Department of Surgery, University of Michigan, Ann Arbor, MI, USA Sayeed Ikramuddin, MD, MHA Department of Surgery, University of Minnesota, Minneapolis, MN, USA Thomas H. Inge, MD, PhD Pediatric Surgery, University of Colorado, Denver and Children’s Hospital Colorado, Aurora, CO, USA Kunoor Jain-Spangler, MD Department of Surgery, Division of Metabolic and Weight Loss Surgery, Duke University, Durham, NC, USA Daniel B. Jones, MD, MS Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA Stephanie B. Jones, MD Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA Gregg H. Jossart, MD, FACS Department of Surgery, California Pacific Medical Center, San Francisco, CA, USA Ashutosh Kaul, MD, FACS, FRCS Department of Surgery, NY Medical College, Valhalla, NY, USA Bariatric Surgery, Greenwich Hospital, Valhalla, NY, USA Minimally Invasive Surgery, Westchester Medical Center, Valhalla, NY, USA Jason W. Kempenich, MD, FACS Department of Surgery, UT Health San Antonio, San Antonio, TX, USA Keith Chae Kim, MD, FACS Department of General Surgery/Bariatrics, Center for Metabolic and Obesity Surgery at Advent Health Celebration, Celebration, FL, USA xxvi Neil A. King, MD Garlock Division of Surgery, Mount Sinai Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA Michelle Cordoba Kissee, MD Endocrinology, Diabetes, Metabolism and Bariatrics, DHR Health System, University of Texas Rio Grande Valley, Edinburg, TX, USA Riley Katsuki Kitamura, MD General Surgery, Palo Alto VA Health Care System, Palo Alto, CA, USA Shanu N. Kothari, MD Minimally Invasive Bariatric Surgery, Department of General Surgery, Gundersen Health System, La Crosse, WI, USA Abraham Krikhely, MD Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA Cindy M. Ku, MD Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA Marina Kurian, MD Department of Surgery, NYU School of Medicine, New York, NY, USA Hendrikus J. M. Lemmens, MD, PhD Multispecialty Division, Department of Anesthesia, Stanford University School of Medicine, Stanford, CA, USA Robert B. Lim, MD Minimally Invasive Surgery, Department of Surgery, Tripler Army Medical Center, Honolulu, HI, USA Zeyad Loubnan, MD Department of Surgery, NYU School of Medicine, New York, NY, USA Rami E. Lutfi, MD, FACS, FASMBS Department of Surgery, University of Illinois at Chicago, Chicago, IL, USA Pearl K. Ma, MD Department of Surgery, Minimally Invasive and Bariatric Surgery, Fresno Heart and Surgical Hospital, University of California San Francisco, Fresno, CA, USA Eric Marcotte, MD, FACS, FASMBS Department of Surgery, Division of GI/Minimally Invasive Surgery, Loyola University Medical Center, Stritch School of Medicine, Maywood, IL, USA Samer G. Mattar, MBBCh, FRCS, FACS, FASMBS Swedish Medical Center, Seattle, WA, USA Corrigan L. McBride, MD, MBA Bariatric Services, Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA Emanuele Lo Menzo, MD, PhD, FACS, FASMBS Department of General Surgery, The Bariatric and Metabolic Institute, Cleveland Clinic Florida, Weston, FL, USA Marc P. Michalsky, MD Clinical Surgery and Pediatrics, Center for Healthy Weight & Nutrition, Pediatric Surgery Department, Nationwide Children’s Hospital, Columbus, OH, USA Dean J. Mikami, MD Department of Surgery, University of Hawaii John A. Burns School of Medicine, Honolulu, HI, USA Tripurari Mishra, MD Minimally Invasive Bariatric Surgery and Advanced Laparoscopy Fellowship, Gundersen Medical Foundation, La Crosse, WI, USA Rachel L. Moore, MD, FACS, FASMBS Moore Metabolics & Tulane University Department of Surgery, New Orleans, LA, USA John M. Morton, MD, MPH, FACS, FASMBS, ABOM Bariatric and Minimally Invasive Surgery Division, Yale School of Medicine, New Haven, CT, USA Contributors Contributors xxvii Geoffrey S. Nadzam, MD, FACS, FASMBS Department of Surgery, Yale School of Medicine, New Haven, CT, USA Estefano A. Negri, MD The Center for Obesity and Diabetes, Oswaldo Cruz German Hospital, São Paulo, Brazil Lucia H. Nguyen, MD General Surgery, Baylor Scott and White Health, Temple, TX, USA Ninh T. Nguyen, MD, FASMBS Department of Surgery, Division of Gastrointestinal Surgery, University of California, Irvine Medical Center, Orange, CA, USA Brittany Nowak, MD General Surgery, NYU Langone Medical Center, New York, NY, USA Nabeel R. Obeid, MD Department of Surgery, University of Michigan, Ann Arbor, MI, USA J. Patrick O’Leary, MD Department of Surgery, Florida International University, Herbert Wertheim College of Medicine, Miami, FL, USA Robert W. O’Rourke, MD Department of Surgery, University of Michigan, Ann Arbor, MI, USA Bariatric Surgery Program, Division of General Surgery, Ann Arbor Veterans Administration Hospital, Ann Arbor, MI, USA Manish Parikh, MD Bariatric Surgery, Bellevue Hospital Center, NYU Langone Medical Center, New York, NY, USA Richard M. Peterson, MD, MPH, FACS, FASMBS Bariatric and Metabolic Surgery, Department of Surgery, UT Health San Antonio, San Antonio, TX, USA Anthony T. Petrick, MD, FACS, FASMBS Geisinger Medical Center, Danville, PA, USA Thomas P. Petrick, MD Department of Surgery, Chinle Comprehensive Health Care Facility, Chinle, AZ, USA Tarissa Z. Petry, MD The Center for Obesity and Diabetes, Oswaldo Cruz German Hospital, São Paulo, Brazil Alfons Pomp, MD, FRCSC, FACS, FASMS GI Metabolic and Bariatric Surgery, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY, USA Jaime Ponce, MD, FACS, FASMBS Bariatric Surgery, Metabolic and Bariatric Care, CHI Memorial Hospital, Chattanooga, TN, USA David A. Provost, MD, FASMBS, FACS Division of General and Bariatric Surgery, Baylor Scott & White Medical Center, Temple, TX, USA Aurora D. Pryor, MD, FACS Department of Surgery, Stony Brook Medicine, Stony Brook, NY, USA Kevin M. Reavis, MD, FACS Foregut and Bariatric Surgery, Division of Gastrointestinal and Minimally Invasive Surgery, The Oregon Clinic, Legacy Weight and Diabetes Institute, Portland, OR, USA Christine Ren-Fielding, MD, FACS, FASMBS General Surgery, Division of Bariatric Surgery, NYU Langone Medical Center, New York, NY, USA Jaime A. Rodriguez, MD Department of Surgery, Florida International University, Herbert Wertheim College of Medicine, Miami, FL, USA Ann M. Rogers, MD Penn State Surgical Weight Loss Program, Department of Surgery, Penn State Hershey Medical Center, Hershey, PA, USA xxviii Raul J. Rosenthal, MD, FACS, FASMBS Department of General Surgery, The Bariatric and Metabolic Institute, Cleveland Clinic Florida, Weston, FL, USA Francesco Rubino, MD Bariatric and Metabolic Surgery, King’s College London Consultant, King’s College Hospital, London, UK Michelle H. Scerbo, MD, MS Department of Surgery, Division of Minimally Invasive and Elective General Surgery, McGovern Medical School, UT Health, Houston, TX, USA Keith Scharf, DO, FACS, FASMBS Bariatric Surgery, Department of General Surgery, Loma Linda University Health, Loma Linda, CA, USA Philip R. Schauer, MD Department of General Surgery, Bariatric and Metabolic Institute, Cleveland Clinic, Cleveland, OH, USA Federico J. Serrot, MD Section of Minimally Invasive Surgery, Department of General Surgery, The Bariatric and Metabolic Institute, Cleveland Clinic Florida, Weston, FL, USA Shinil K. Shah, DO Department of Surgery, Division of Minimally Invasive and Elective General Surgery, McGovern Medical School, UT Health, Houston, TX, USA Michael E. DeBakey Institute for Comparative Cardiovascular Science and Biomedical Devices, Texas A&M University, College Station, TX, USA Adam C. Sheka, MD Department of Surgery, University of Minnesota, Minneapolis, MN, USA Ranjan Sudan, MD Department of Surgery, Division of Metabolic and Weight Loss Surgery, Duke University, Durham, NC, USA Amit Surve, MD Bariatric Medicine Institute, Salt Lake City, UT, USA William A. Sweet, MD, FACS (Retired) Reading Hospital and Medical Center, West Reading, PA, USA Patrick J. Sweigert, MD Department of Surgery, Loyola University Medical Center, Maywood, IL, USA Samuel Szomstein, MD, FACS, FASMBS Department of General Surgery, The Bariatric and Metabolic Institute, Cleveland Clinic Florida, Weston, FL, USA Dana A. Telem, MD, MPH Clinical Affairs, Comprehensive Hernia Program, Department of Surgery, University of Michigan, Ann Arbor, MI, USA Renée M. Tholey, MD Department of Surgery, Thomas Jefferson University Hospital, Philadelphia, PA, USA David S. Tichansky, MD Department of Surgery, Thomas Jefferson University Hospital, Philadelphia, PA, USA Hussna Wakily, MD Division of General Surgery, Department of Surgery, NY and NJ Surgical Associates, Queens, NY, USA Elizaveta Walker, MPH Department of Surgery, Oregon Health & Science University, Portland, OR, USA D. Brandon Williams, MD Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA Erik B. Wilson, MD Department of Surgery, Division of Minimally Invasive and Elective General Surgery, McGovern Medical School, UT Health, Houston, TX, USA Contributors Contributors xxix Keith M. Wirth, MD Department of Surgery, University of Minnesota, Minneapolis, MN, USA Bruce M. Wolfe, MD Department of Surgery, Oregon Health & Science University, Portland, OR, USA Hinali Zaveri, MD Bariatric Medicine Institute, Salt Lake City, UT, USA Natan Zundel, MD Department of Surgery, Florida International University Herbert Wertheim College of Medicine, North Miami Beach, FL, USA Part I Basic Considerations 1 Epidemiology and Discrimination in Obesity R. Armour Forse, Monica M. Betancourt-Garcia, and Michelle Cordoba Kissee Chapter Objectives At the end of the chapter, the reader should be able to describe: 1. Definition of obesity 2. Epidemiology of obesity • Global burden of obesity • Obesity in the United States 3. Disparities in obesity by age, race, ethnicity, gender, and socioeconomic status 4. Obesity-related discrimination, especially in spheres of employment, health care, and children 5. Effect of discrimination on obese individuals Introduction Obesity has become one of the most significant public health challenges in both economically developed and developing regions of the world. In 2016, more than 1.9 billion adults worldwide were overweight, and, of these, more than 650 million were obese, a number that has tripled since the 1970s [1]. There are an estimated 2.5 people added to the global population each second, and one of them will be obese or R. A. Forse (*) DHR Health Institute for Education and Innovation, DHR Health System, Department of Surgery, University of Texas Rio Grande Valley, Edinburg, TX, USA e-mail: r.forse@dhr-rgv.com M. M. Betancourt-Garcia DHR Health Institute for Research and Development, DHR Health System, Edinburg, TX, USA M. C. Kissee Endocrinology, Diabetes, Metabolism and Bariatrics, DHR Health System, University of Texas Rio Grande Valley, Edinburg, TX, USA overweight. In the United States it is estimated that 39.8% of the adult population is obese [2]. Obesity is associated with markedly reduced life expectancy, thus becoming a leading cause of preventable deaths in the United States. In 2015, high body mass index (BMI) contributed to 4 million deaths and contributed to 120 million disability-adjusted life-years (Fig. 1.1). High BMI has been shown to be associated with hypertension, hyperlipidemia, coronary artery disease, abnormal glucose tolerance or diabetes, sleep apnea, nonalcoholic fatty liver disease, musculoskeletal disorders, kidney disease, and certain cancers including esophageal, pancreatic, renal cell, postmenopausal breast, endometrial, cervical, and prostate cancers [3]. Health-care costs associated with obesity are high and climbing. In 1998, the estimated annual cost of obesity was $78.5 billion. Today, in the United States alone, obesity and its related illnesses cost an estimated $191 billion with the medical costs $2741 higher, annually, for people who are obese than those of normal weight [4]. Social, psychological, and economic consequences are also well-recognized. A large amount of research is focused on the understanding of obesity, and many public health efforts have been directed toward controlling its exponential growth. Definition of Obesity The World Health Organization (WHO) defines obesity as a condition of excessive fat accumulation in the body to the extent that health and well-being are adversely affected [1]. If the amount of body fat exceeds normal physiological values, a person is obese. Although this definition appears simple on the surface, it has major limitations. The physiologically normal amount of body fat depends on age and on gender with high variation among individuals. Newborns have 10–15% body fat (BF), and during the 1st year of life, this increases to about 25%. After that, BF% slowly decreases again to 15% of body weight at the age of 10 years, when differences between the sexes become more apparent. During © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_1 3 4 R. A. Forse et al. Musculoskeletal Disorders Cardiovascular Diseases a Disability-Adjusted Life-Years in 1990 Cancers Chronic kidney Disease Diabetes Mellitus b Disability-Adjusted Life-Years in 2015 10 10 8 8 6 Disability-Adjusted Life-Years (in millions) Disability-Adjusted Life-Years (in millions) 1.6% 1.6% 2.1% 4 25.6% 35.4% 0.3% 2 0.6% 0.8% 0.3% 20 6 18.7% 25 3.3% 0.2% 2.5% 0.4% 6.4% 0.8% 0.3% 2.8% 11.5% 2.1% 0 3.1% 30 35 40 45 50 4.2% 2.9% 3.4% 5.8% 33.7% 4 2 4.1% 0 2.8% 20 25 18.0% 4.7% 30 35 40 45 50 Body-Mass Index Body-Mass Index Fig. 1.1 Global disability-adjusted life-years associated with a high body mass index (1990–2015). Shown are the number of global disability-­adjusted life-years (in millions) related to a high body mass index (BMI) among adults according to the cause and the level of BMI in 1990 (Panel a) and in 2015 (Panel b). (Source: GBD 2015 Obesity Collaborators [3]. Reprinted with permission from Massachusetts Medical Society) sexual maturation, girls experience an increase in their body fat again, up to 25%, whereas boys keep about the same BF%. During adulthood, BF% increases slowly with age in both males and females. It is not known whether this age-­ related increase during adult life is a normal physiological effect or whether it is caused by overeating and/or a sedentary lifestyle. Body fat can be measured by different techniques including densitometry, hydrometry, dual energy X-ray absorptiometry (DXA), chemical multi-compartment models, computed tomography (CT), or magnetic resonance imaging (MRI). These methods are not suited for use under clinical conditions and in population-based studies. There are a number of other methods available for large-scale use that can predict body fat (BF)%; these include skinfold thickness measurements, bioelectrical impedance, the use of body mass index (BMI) and waist circumference, and the more recently described body adiposity index (BAI) [5]: divided by height in m2. Generally, healthy BMI range is from 18.5 to 24.9 kg/m2. Overweight is defined as a BMI from 25 to 29.9 kg/m2, and obesity is defined as a BMI of 30 kg/m2 or greater. Obesity can further be subdivided based on subclasses of BMI, as shown in Table 1.1 [6, 7]. Extreme obesity is defined as a BMI greater than 40 kg/m2. Waist circumference can also be used in combination with a BMI value to evaluate health risk for individuals. The waist/hip ratio relates to the distribution of body fat. Patients with a waist/hip ratio of less than one tend to have more of a peripheral fat distribution ratio often referred to as having a “pear” Table 1.1 Categories of BMI and disease risk relative to normal weight and waist circumference BAI = ( hip circumference / height1.5 ) - 18. These methods rely on statistical relationships between easily measurable parameters and a method of reference, normally densitometry, deuterium oxide dilution, or DXA. As the range of BF% varies largely and is dependent on age and sex, clearly defined cutoff points for obesity, expressed as BF%, cannot easily be established. There is no doubt that these clinical measures are limited in terms of accuracy, but they are very portable and applicable and give meaningful trends when used over time. Of the aforementioned parameters, the one that is most widely applied is BMI, which is determined by weight Underweight Normala Overweight Obesity Obesity Extreme obesity Relative disease risk by waist circumference (type 2 diabetes, hypertension, cardiovascular disease) Men >102 cm Men ≤102 cm (>40 in) (≤40 in) Women Obesity Women ≤88 cm >88 cm (>35 in) (≤35 in) BMI kg/m2 class <18.5 – – – 18.5–24.9 – – – 25.0–29.9 – Increased High 30.0–34.9 I High Very high 35.0–39.9 II Very high Very high III Extremely high Extremely ≥40 high Modified from [6, 7] a Increased waist circumference can also be a marker for increased risk even in persons of normal weight 1 Epidemiology and Discrimination in Obesity 5 distribution. This fat distribution has lower health risks. Patients with a waist/hip ratio of greater than one are referred to as having an “apple” or central fat distribution, and these patients are considered to have a high health risk [8]. In children (2–19 years of age), overweight is defined as a BMI-for-­ age greater than or equal to the 85th percentile and less than the 95th percentile on the Centers for Disease Control and Prevention (CDC) growth charts [9]. Obesity is defined as a BMI-for-age greater than or equal to the 95th percentile on the CDC growth charts. It is well accepted that BMI is an estimate rather than an accurate measurement. It fails to account for fitness, and there is a wide variation of body adiposity in the same BMI range. In general, adiposity has been shown to vary among men and women (with women having more adiposity for the same BMI group) and across different age groups (adiposity increases with age). It has also been noted that in the same BMI range, Asians and African-Americans have more prevalence of diseases such as hypertension and diabetes. Using BMI as the only qualifying requirement for bariatric surgery runs the risk of discriminating against these groups, and care may be denied to patients who may benefit from if delivery of care is based upon this imperfect and somewhat arbitrary measure of obesity. Epidemiology of Obesity Global Burden of Obesity Overweight and obesity are increasing public health challenges in both economically developed and developing regions of the world, with nearly one third of the world’s population (1.9 billion adults and children) overweight or obese [1] (Fig. 1.2). This is a substantial increase from 1980 when only 5% of men and 8% of women were obese [10]. It Male Female Global revalence of Overweight and Obesity P in the United States The strongest data on obesity prevalence rates over time in the United States come from the results of the National Low SDI Low-middle SDI Middle SDI High-middle SDI High SDI 2010 2015 C Obesity in Adults According to Year 20 15 Prevalence (%) Fig. 1.2 Prevalence of obesity at the global level, according to sociodemographic index (SDI). Age standardized prevalence trends at the global level and according to SDI quintile from 1980 to 2015 among adults. (Source: GBD 2015 Obesity Collaborators [3]. Reprinted with permission from Massachusetts Medical Society) is estimated that if recent trends continue, by 2030 up to 57.8% of the world’s adult population (3.3 billion people) could be either overweight or obese [11]. In general, the prevalence of overweight and obesity is higher in economically developed countries; however, the prevalence has continuously increased across all levels of development, regardless of economic growth [3, 11]. Approximately 604 million adults are obese, and the global prevalence is higher in women than in men [3]. More than 50% of the world’s obese population live in only ten countries, including the United States, China, India, Russia, Brazil, Mexico, Egypt, Germany, Pakistan, and Indonesia [12]. Obesity has been thought of as a high-income country problem, but the prevalence of overweight and obesity is also on the rise in developing countries, particularly in urban settings. This is in part fueled by industrialization, the creation of mass transit systems, lifestyle changes, and the promotion of unhealthy “fast foods” in these countries in the last two decades. Many developing nations are now facing a “double burden” of disease as is seen in much of Asia, Latin America, the Middle East, and Africa. While they continue to deal with the problems of infectious disease and undernutrition, they are experiencing a rapid upsurge in noncommunicable disease risk factors such as obesity and overweight. It is not uncommon to find undernutrition and obesity existing side by side in the same community and the same household. Conversely, accelerated growth rates in overweight and obesity seen from 1992 to 2002 have slowed in developed countries [12]. 10 5 0 1980 1985 1990 1995 2000 Year 2005 6 R. A. Forse et al. Health and Nutrition Examination Surveys (NHANES). The NHANES program of the National Center for Health Statistics (NCHS), Centers for Disease Control and Prevention, includes a series of cross-sectional nationally representative health examination surveys beginning in 1960 [13–17]. In each survey, a nationally representative sample of the US civilian non-institutionalized population was selected using a complex, stratified, multistage probability cluster sampling design. In the 2009–2010 and 2015–2016 surveys, household interview and a physical examination were conducted for each survey participant including height and weight measured as part of a comprehensive set of body measurements by trained health technicians, using standardized measuring procedures and equipment excluding pregnant women and persons missing a valid height or weight measurement [2, 18]. In the 2009–2010 survey, age was based on age at the time of interview and grouped into 20–39 years of age, 40–59 years of age, and 60 years and older. Race and ethnicity were self-reported and for purposes of this report were classified as non-Hispanic white, non-­ Hispanic black, Mexican-American, other Hispanic, and others. Results from the 2009–2010 National Health and Nutrition Examination Survey (NHANES), using measured heights and weights, indicate that an estimated 33.0% of US adults aged 20 and over are overweight, 35.7% are obese, and 6.3% are extremely obese [18]. In 2009–2010, the age-­ adjusted mean BMI was 28.7 (95% CI, 28.3–29.1) for men and also 28.7 (95% CI, 28.4–29.0) for women [18]. Figure 1.3 shows the trends in overweight and obesity among adults from 1960 to 2010 [16]. In this figure it is interesting to note that the rate of overweight has been more or less stable, but there have been significant increases in the rates of obesity, with obesity rates having very recently overtaken the rate of prevalence of overweight in the adult population. The National Center for Health Statistics analyzed obesity in an updated 2015–2016 study. The participants in the 2015–2016 survey were grouped into 20–39 years of age, 40–59 years of age, and 60 years and older. Race and ethnicity were classified as non-Hispanic white, non-Hispanic black, non-Hispanic Asian, and Hispanic. The data for adults suggests a steady prevalence of obesity from the 1960s through the 1980s, with a steady increase in obesity since the late 1980s and today in the United States, with the estimated age-adjusted prevalence moving upward from a previous level of 23.0% in 1988–1994 to approximately 39.8% in 2015–2016 [2, 16]. The age-adjusted prevalence of obesity was 37.9% among adult men and 41.1% among adult women [2]. The highest prevalence of obesity was seen in the 40–59 age group for both men and women. One of the national health objectives for the Healthy People 2020 initiative is to reduce the prevalence of obesity among adults by 10–30.5% [19]. Figure 1.4 depicts the trends in obesity prevalence among adults and youth from 1999 to 2016. Data suggest that those who are obese may be gaining weight at a more rapid pace than those who are not. Data from the Behavioral Risk Factor Surveillance System (a random-­digit telephone survey of the household population of the United States) shows that it is not just that more Americans are becoming obese but that it is the most severe obesity that is increasing the most in relative terms. From 2000 through 2005, the prevalence of obesity (self-reported) increased by 24%, the prevalence of a self-reported BMI greater than 40 increased by 50%, and the prevalence of a BMI greater than 50 increased by 75% [20]. The greatest relative increase has been in the proportion of individuals with a BMI greater than 50 kg/m2. The most recent NHANES data also confirm this trend: the percentage of the population with a BMI greater than 40 has increased from 0.9% in the 1960s to approximately 6% at the current time [17]. 60 Percent 50 40 Overweight 30 20 10 0 Obese Extremely obese 19601962 19711974 19761980 Fig. 1.3 Increasing trends in overweight, obesity, and extreme obesity among US men ages 20–74 years, spanning the years 1960–1962 through 2009–2010. Age adjusted by the direct method to the 2000 US Census population using age groups 20–39, 40–59, and 60–74. Overweight is a body mass index (BMI) of 25 kg/m2 or greater but less than 30 kg/m2. Obesity is a BMI greater than or equal to 30 kg/m2. 19881994 2001- 2005- 20092002 2006 2010 Extreme obesity is a BMI greater than or equal to 40 kg/m2. (Data sources: CDC/NCHS, National Health Examination Survey I 1960– 1962; National Health and Nutrition Examination Survey (NHANES) I 1971–1974; NHANES II 1976–1980; NHANES III 1988–1994; NHANES 1999–2000, 2001–2002, 2003–2004, 2005–2006, 2007– 2008, and 2009–2010. Data from Fryar et al. [16]) 1 Epidemiology and Discrimination in Obesity Fig. 1.4 Trends in obesity prevalence among adults aged 20 and over (age adjusted) and youth aged 2–19 years: United States, 1999–2000 through 2015–2016. (Source: Hales et al. [2]) 7 40 Percent 30 37.7 30.5 30.5 32.2 34.3 35.7 34.9 16.8 16.9 16.9 17.2 2007– 2008 2009– 2010 2011– 2012 2013– 2014 33.7 39.6 Adults1 20 13.9 15.4 17.1 15.4 18.5 Youth1 10 0 1999– 2000 2001– 2002 2003– 2004 2005– 2006 2015– 2016 Survey years Epidemiology of Childhood Obesity sity was seen at all race, ethnicity, and income levels [15], those of Hispanic origin and non-Hispanic black children There were more than 213 million children aged 5–19 years had a higher prevalence compared to non-Hispanic white old that were overweight and 125 million children with obe- and non-Hispanic Asian children [2]. sity as of 2016 [21]. In a study that included data from 200 Overweight children and adolescents are more likely to countries and territories in all regions of the world, obesity be overweight or obese adults. Those with a BMI between prevalence increased in every region. Rates rose from 0.7% the 75th and 84th percentiles were up to 20 times more likely to 5.6% in girls and from 0.9% to 7.8% in boys since 1975 than their leaner counterparts of becoming overweight or [21], and the rates of increase were highest in early adult- obese as young adults [23]. Childhood obesity can lead to hood between the ages of 15 and 24 years old [30]. The high- immediate psychosocial and metabolic problems but may est global standardized mean BMI in children and adolescents also have lasting effects on health, especially if this weight is were found in high-income English-speaking regions carried into adulthood. A complex interaction of genetic fac(22.4 kg/m2) and Polynesia (23.1 kg/m2) [21]. tors such as variation in DNA sequence or expression and Children in developing countries are more vulnerable to epigenetic factors including in utero environment, behavior, inadequate prenatal, infant, and young child nutrition. At the lifestyle, ethnic variability in body composition, and values/ same time, they are exposed to high-fat, high-sugar, high-­ perceptions has led to an increase in the prevalence of obesalt, energy-dense, micronutrient-poor foods, which tend to sity and its assocaited chronic diseases such as high blood be lower in cost. Additionally, urbanization and mechaniza- pressure, high cholesterol, diabetes, cardiovascular disease, tion, higher rates of television viewing and electronic device and many cancers. use, and increasing pressure among children in developing countries to perform scholastically have led to a sharp decline in physical activity. Interaction of these factors with chang- Obesity and Age ing dietary patterns results in drastic increases in childhood Obesity rates are high in most age groups. For both men and obesity and metabolic syndrome. In the United States in 2015–2016, the prevalence of obe- women, the prevalence of obesity is higher among middle-­ sity in children 2–19 years of age was 18.5% and affected aged adults (42.8%) between 40 and 59 years of age than about 13.7 million children and adolescents, with the highest younger adults 20 and 39 years of age (35.7%) [2]. The rate prevalence among adolescents 12–20 years of age [22] of increase, however, is higher in early adulthood between 15 (Fig. 1.5). The prevalence of obesity decreased with increas- and 24 years of age [3]. Obesity rates, in general, increase ing level of education and income in the household ranging with age until approximately 75 years of age, when rates from 18.9% among children and adolescents in the lowest decline. The decline in obesity rates in the elderly could be income group and 10.9% among those in the highest income attributed to a decrease in lean body mass and a tendency to group [2]. While an increase in prevalence of childhood obe- gain fat in the older patient, which plateaus as the older 8 R. A. Forse et al. Fig. 1.5 Prevalence of obesity among youth aged 2–19 years, by sex and race and Hispanic origin: United States, 2015–2016. (Source: Hales et al. [2]) Non-Hispanic white 30 Non-Hispanic black Hispanic Non-Hispanic Asian 28.0 1–3 25.8 1,2 25.1 1,2 25 1,2 23.6 22.0 1,2 Percent 20 15 19.0 14.6 14.1 11.0 13.5 11.7 10.1 10 5 0 Total patient establishes a new weight set point. In addition, there is increasing mortality from obesity-related conditions with age; a significantly higher all-cause mortality has been noted in obese individuals compared to normal weight subjects, with one study predicting that mortality was likely to occur 9.44 years earlier for those who were obese (BMI, ≥30) [24]. Racial, Ethnic, and Income Disparities Increasing BMI and increasing obesity prevalence are affecting the entire adult population with no group being immune [25, 26]. Increasing rates of obesity are seen across men and women of all ethnic groups, of all ages, and of all educational and socioeconomic levels. Still, racial, ethnic, and socioeconomic disparities are seen in the prevalence of obesity, and some subgroups in the population are affected to a greater extent than others. Boys Girls the 12-year period from 1999 through 2010 but has slowed in recent years [2]. For women, the age-adjusted prevalence is 41.1%, and the range is from 14.8% among non-Hispanic Asian women to 54.8% among non-Hispanic black women, and a prevalence of 50.6% in Hispanics (Fig. 1.6) [2]. Within all race/ethnicity groups, women have a higher prevalence than men. Interestingly, non-Hispanic black women also experienced a decline in prevalence since the 2011 survey from 56.9% to 54.8% [2]. These disparities are likely a combination of differences in socioeconomic status, environmental and social risk factors among racial groups, and genetic, epigenetic, and metabolic factors, which suggest a hereditary nature of disease and similar obesity patterns in families [14, 15]. Obesity and Socioeconomic Status The prevalence of overweight and obesity in both adults and children is higher in economic developed countries [11]. However, when dividing countries into quintiles of economic There are significant racial and ethnic disparities in obesity status, the most rapid relative increase in obesity occurred in prevalence among US adults. Among men, overall age-­ individuals living in countries with low and low-middle adjusted obesity prevalence is 37.9%, but within race/ethnic- sociodemographic characteristics [3]. ity groups, prevalence ranges from 10.1% among On an individual level, obesity prevalence decreases as non-Hispanic Asian men to 43.1% among Hispanic men income increases [14] with obesity among adults approxi(Fig. 1.6). Between the 1988–1994 and 2015–2016 surveys, mately 8% lower in the highest income group (>350% of the prevalence of obesity among men increased, from 20.3% the federal poverty level) compared to those living on less to 37.9% in non-Hispanic white men and 23.9–43.1% among than 130% of the federal poverty level. The largest contrast Hispanic men [2]. Prevalence for non-Hispanic black men in prevalence is observed between women with income rose from 21.1% in 1988–1994 to 37.3% in 2007–2008 but >350% of the federal poverty level and those 130% below has since dropped slightly to 36.9%. For men, the overall the federal poverty level, 29.7% versus 45.2%, prevalence of obesity showed a significant linear trend over respectively. Obesity and Race 1 Epidemiology and Discrimination in Obesity Fig. 1.6 Age-adjusted prevalence of obesity among adults aged 20 and over, by sex and race and Hispanic origin: United States, 2015–2016. (Source: Hales et al. [2]) 9 Non-Hispanic white 60 Non-Hispanic black Hispanic Non-Hispanic Asian 54.8 1,2 1,2 50 46.8 50.6 47.0 1,2 1,2 43.1 1,4 Percent 40 1 37.9 1 37.9 1,3,436.9 38.0 1 30 20 12.7 10 14.8 10.1 4 0 Total Education has a large influence on obesity rates. Age-­ adjusted prevalence of obesity among college graduates is 27.8%, while prevalence for those who were high school graduates or less is 40.0% [27]. It is interesting to note that some of these trends differ when considering race and ethnicity. For instance, women in general have higher obesity rates than do men regardless of income or education level, except for college-educated high-­ income non-Hispanic white men who have higher obesity rates than their non-Hispanic white female counterparts. Non-Hispanic Asian individuals have prevalence rates at approximately 11%, and this remains consistent for both men and women regardless of income or education level [27]. Trends in Obesity Close examination of trends in obesity shows that this epidemic arose from gradual yearly weight gain in the population produced from a slight consistent degree of positive energy balance (i.e., energy intake exceeding energy expenditure). Using longitudinal and cross-sectional data sets, it was found that the average adult in the United States has gained an average of 1–2 lb/year for the past two to three decades [28]. Assuming that an excess of 3500 kcal produces 1 lb of weight gain and assuming that excess energy was stored with an efficiency of 50%, that weight gain in 90% of the adult population is attributable to a positive energy balance of approximately 100 kcal/day. Thus, it seems that the obesity epidemic arose gradually over a long period because of a slight but consistent degree of positive energy balance. Our bodies are designed to work best in a setting in which food was inconsistent and high levels of physical activity Men Women were required to secure food and shelter and for transportation. In previous times, this biology was adequate to allow most people to maintain a healthy weight without conscious effort. Body weight regulation was achieved for most with simple physiological control. The situation is different in today’s environment. Securing food and shelter and moving around no longer requires the high levels of physical activity needed in the past. Technology has made it possible to be productive while being largely sedentary. Advancements in workplace technology and reduction of manual labor have resulted in decreased energy expenditure. Factors such as urban design, land use, public transportation availability, density and location of food stores and restaurants, and neighborhood barriers such as safety and walkability contribute to unhealthy lifestyles. Significant changes have taken place in the food environment with increased accessibility of inexpensive foods. Prices of calorie-dense foods and beverages have decreased considerably in contrast to increasing prices of fresh fruits, vegetables, fish, and dairy items, contributing to increased consumption of unhealthy foods in increasing portion sizes. Significant marketing and advertising of unhealthy, energydense foods by the food industry contribute to excessive food consumption. Under such conditions, weight gain can only be prevented with conscious efforts to eat less eat healthier and/or to be physically active. Obesity rates are increasing among people in all income and educational levels, but absolute rates are lower in those with higher incomes and higher education levels. The finding that minority and low-income individuals are disproportionately affected by obesity is not surprising. The most inexpensive foods are those containing high levels of fat and sugar. The biological preference for these foods combined with easy availability contributes to overeating. Further, minority and low-income individuals may engage in less 10 physical activity than other sectors of the population [29]. One reason for this disparity may be because problems with neighborhood safety in low-income areas may prevent adults and children from engaging in outdoor physical activities. People who have more financial resources combat these circumstances more easily and, consequently, maybe more physically active and less obese than those with fewer resources. Discrimination in Obesity Obesity Discrimination Overweight and obese individuals are vulnerable to negative societal attitudes, stigma, and prejudice. Reported experiences of weight/height discrimination included a variety of settings in major lifetime events and interpersonal relationships. Weight bias has been documented in multiple settings including places of employment, healthcare facilities, educational institutions, mass media, and close interpersonal relationships with friends and family members [30–33]. Obese individuals, especially the severely obese, often confront bias in these various settings. They are frequently the target of ridicule even in their early years in school. Some estimates even suggest that 25% of severely obese women were sexually abused. Obese people have difficulty finding jobs [32], and, when they find employment, they all too often have high levels of absenteeism, poor performance at work, and high health-care costs. Similarly, within their families, the levels of conflict regarding relationships, parenting, and sexuality are high. Data from the two versions of the National Survey of Midlife Development in the United States (MIDUS) have shown increasing incidence of weight-based discrimination from 7% in 1995–1996 to 12% in 2004–2006 [30]. Data for this were drawn from a nationally representative multistage probability sample of community-based English-speaking adults in the contiguous United States. Weight-based discrimination is the third leading cause of prejudice, next only to age and race. In contrast to more widely recognized social stigmas such as gender or race that have legal sanctions in place to protect individuals from discrimination, there are no laws to prohibit weight discrimination, with the exception of a few states. It is unknown how weight discrimination compares in strength or prevalence to discrimination based on these attributes. Because weight stigma remains a socially acceptable form of bias, negative attitudes and stereotypes toward obese persons have been frequently reported by employers, coworkers, teachers, physicians, nurses, medical students, dietitians, psychologists, peers, friends, family members, and even among children aged as young as 3 years [30]. R. A. Forse et al. Employment Discrimination Obesity discrimination is widespread in employment. Employment discrimination may be related to the increasing focus on employee weight and its contribution to employers’ overall costs stemming from increased healthcare costs and decreased productivity from absenteeism, which is perceived among coworkers as laziness and lack of dedication that may lead to heightened discrimination. In 2007, obesity and morbid obesity were associated with an estimated cost of $4.3 billion in the United States with the estimated annual cost of absenteeism being $1026 for a male worker with BMI >40 and $1262 for a female worker with the same BMI [32]. In addition, employing morbidly obese individuals is associated with additional cost to employers; for example, a bariatric chair able to hold 500 lb is estimated to cost $1295, and a bariatric toilet rated at 700 lb is estimated at $1049. Employers have started implementing wellness programs and incentives to control health-care cost. Evidence suggests that medical costs fall by about $3.27 for every dollar spent on wellness programs and that absenteeism costs fall by about $2.73 for every dollar spent [34]. Wellness programs have their own disadvantages; especially those that impose financial risk on unhealthy employees are likely to be regressive because the prevalence of unhealthy conditions typically targeted by wellness programs is highest among people with low socioeconomic status. Data from various surveys consistently document lower labor market participation of overweight and obese individuals. While employers and coworkers may use the data mentioned previously to justify bias against obese workers, this mentality may adversely impact the psychological well-­being of the individual, which in turn can result in lost days at work and decreased productivity. Roehling et al. found that overweight respondents were 12 times more likely, obese respondents were 37 times more likely, and severely obese respondents were 100 times more likely than normal weight respondents to report employment discrimination [32]. In addition, women were 16 times more likely to report weight-related employment discrimination than men [32]. A meta-analysis of 32 experimental studies that investigated weight discrimination in employment settings was recently conducted [31]. Typically, such experimental studies ask participants to evaluate a fictional applicant’s qualifications for a job, where his or her weight has been manipulated (through written vignettes, videos, photographs, or computer morphing). Outcome variables examined in these studies included hiring recommendations, qualification/suitability ratings, disciplinary decisions, salary assignments, placement decisions, and coworker ratings. Across studies, it was demonstrated that overweight job applicants and employees were evaluated 1 Epidemiology and Discrimination in Obesity more negatively and had more negative employment outcomes compared to non-overweight applicants and employees [31]. After adjusting for sociodemographic and health-related variables, an increase in BMI is shown to be associated with a lower percentage of total working years, a lower rate of employment, and a lower probability of regaining employment. Discrimination between normal weight individuals and those overweight or obese has been demonstrated, and there is now evidence that shows society holds very different body standards for men versus women. As a result, the two sexes are discriminated against differently. A 2010 study that compared income earnings across the BMI spectrum found that very thin women are rewarded for being underweight (decrease in weight of 2 standard deviations) and earn approximately $22,000 more than average weight women [3]. However, men who were 2 standard deviations below average resulted in the opposite with an earnings decrement of $17,535 [3]. Overweight and obese women lost roughly between $9000 and $19,000 more than average weight women, but men with an increase in weight of 2 standard deviations actually earned $14,889.00 more than average weight men [3]. Our society has standards of attractiveness that are substantially slimmer for women than men who then consequently experience opposite incentives regarding weight. Overweight women are perceived as fat, lazy, undisciplined, and unsuccessful, while overweight men are perceived as strong, competent, reliable, and tough and are rewarded for gaining weight until the point of obesity. Those that do not resemble the stereotypical gender norms experience financial consequences proportional to their deviation from these norms. Health-Care Discrimination Health-care costs associated with obesity are high, and the estimated annual cost of obesity was $147 billion in 2008 with the medical costs for people who are obese being $1429 higher annually than those of normal weight in the same year [35]. Studies demonstrate negative stereotypes and attitudes toward obese patients by a range of health-care providers and fitness professionals [33]. There is also research indicating that providers spend less time in appointments and provide less health education with obese patients compared with thinner patients. There is evidence indicating that these attitudes begin during medical school. Medical students perceive overweight and obese patients as ugly, lazy, sloppy, depressed, and a main target of derogatory humor in the hospital setting [36]. In response, obese individuals frequently report experiences of weight bias in health care. Obese patients also indicate that they feel disrespected by providers, perceive that they will not be taken seriously because of 11 their weight, report that their weight is blamed for all of their medical problems, and are reluctant to address their weight concerns with providers. A number of studies demonstrate that obese persons are less likely to undergo age-appropriate preventive cancer screenings. Lower rates of preventive health care exist even after control for factors such as less education, lower income, lack of health insurance, and greater illness burden. In a survey of obese women about their perceived barriers to routine gynecological cancer screenings, weight was reported to be a major barrier to seeking health care with perception of disrespectful treatment and negative attitudes from providers, embarrassment about being weighed, and receiving unsolicited advice to lose weight, and it was also reported that gowns, examination tables, and other medical equipment were too small to be functional for their body size [37]. Obesity Discrimination in Children The psychological and social consequences of being overweight or obese are likewise experienced by children. Obesity stigmatization and discrimination occurring in adulthood actually emerges in children as early as 2 years old [40]. Children can be victims of physical (e.g., kicking, pushing, hitting), verbal (e.g., being teased, name calling, derogatory remarks), or relational (e.g., being ignored or avoided, social exclusion, being targets of rumors) aggression [41]. Studies have shown that preadolescent obese boys and girls are more likely to be victims of bullying because they deviate from appearance ideals [42]. Overweight and obese peers are associated with negative adjectives and less preferred as friends than the normal weight children, and the prejudice was stronger toward females than males [43]. Interestingly, these attitudes persist despite the child’s own body size. In fact, overweight children have demonstrated stronger stigmatism than those who are not overweight [44]. There are two studies that suggest even parents have biases against their own children with obesity by not providing the same support emotionally and financially than would be given to average weight children [45, 46]. Furthermore, these prejudices follow them through adulthood and can have major social impacts. One survey found that a few extra pounds could reduce a woman’s chance of getting married by 20% [47]. This study also noted overweight females completed fewer years of school, had lower household incomes, and had 10% higher rates of household poverty [47]. It is clear that a child’s perception of an endomorph is established early in childhood and discrimination of these individuals begins early in life, especially in girls. 12 Effect of Discrimination on Obese Individuals Perceived weight stigma and discrimination have a vast impact on the quality of life of overweight individuals [30]. A number of studies have consistently demonstrated that experiencing weight stigma increases the likelihood of engaging in unhealthy eating behaviors and lower levels of physical activity, both of which exacerbate weight gain and obesity. For instance, as a result of discrimination in the sphere of health care, overweight patients might be reluctant to seek medical care, be more likely to cancel or delay medical appointments, or put off important preventative ­ health-­care services [33]. besity Discrimination and Psychological O and Physical Health Emerging research suggests that weight stigma invokes psychological stress, which contributes to poor physical health outcomes for obese individuals and is a risk factor for depression, anxiety, low self-esteem, and body dissatisfaction [31]. Adults who experience weight-based stigmatization engage in more frequent binge eating, are at increased risk for maladaptive eating patterns, and are more likely to have a diagnosis of binge eating disorder. Obesity has been associated with impaired quality of life. A 1997 study measured the impact of obesity on functional health status and subjective well-being. Health-related quality of life, as measured by the Medical Outcomes Study Short Form-36 Health Survey, of more than 300 obese persons seeking treatment for obesity at a university-based weight management center was compared with that of the general population and with that of other patients with chronic medical conditions [38]. Obese participants (mean BMI of 38.1) reported significantly lower scores (more impairment) on all eight quality-of-life domains, especially bodily pain and vitality. The morbidly obese (mean BMI of 48.7) reported significantly worse physical, social, and role functioning; worse perceived general health; and greater bodily pain than did the mildly obese (mean BMI of 29.2) or moderately to severely obese (mean BMI of 34.5) [38]. The obese participants also reported significantly greater disability attributable to bodily pain than did participants with other chronic medical conditions. There is also emerging evidence for physiological effects, with weight discrimination having been shown to be related to increased blood pressure, chronic inflammation, greater disease burden, and even increased risk of mortality [39]. There is a common perception that weight discrimination might encourage individuals with obesity to lose weight, but research suggests the opposite to be true. Both adults and children who experience weight-related discrimination are R. A. Forse et al. actually more likely to engage in behaviors that promote the progression of obesity, including disordered eating, poor adherence to healthy food choices, increased energy intake, and avoidance of physical activity, thus further increasing the prevalence and severity of obesity through a vicious cycle of weight gain and discrimination [39]. Obesity Discrimination and Public Health It has been shown that the more a disease is perceived as under volitional control, the more stigmatizing it is—with obesity generally being perceived as highly volitional. Numerous studies have documented harmful weight-based stereotypes that overweight and obese individuals are lazy, weak-willed, unsuccessful, and unintelligent, lack self-­ discipline, have poor willpower, and are noncompliant with weight loss treatment. Society regularly regards obese persons as architects of their own ill-health, personally responsible for their weight problems because of laziness and overeating. Because of these common perceptions, weight stigmatization is regarded as justifiable (and perhaps necessary) because obese individuals are believed to be personally responsible for their weight and that stigma might even serve as a useful tool to motivate obese persons to adopt healthier lifestyle behaviors, with weight stigma being suggested by some as a method for obesity control [39]. Although assumptions about personal responsibility in obesity and justification of weight stigma are prevalent in our national mindset, considerable scientific evidence has emerged to challenge them. Many significant contributors to obesity are beyond the control of individuals. In addition to the important role of genetic and biological factors regulating body weight, multiple social and economic influences have significantly altered the environment to promote and reinforce obesity. It has been shown that most behavioral and dietary interventions produce a modest 10% weight loss with a high rate of weight regain. This is associated with improvements in obesity-related health consequences such as diabetes, hypertension, and cardiovascular disease but is unlikely to alter appearance or translate into a non-obese BMI and is unlikely to be significant to reduce obesity-related stigma and discrimination. Obesity stigma creates significant barriers in efforts to address the epidemic. Current public health approaches to tackling this epidemic focus on providing education to the affected individuals rather than providing a comprehensive plan to tackle this epidemic. This approach is based on the assumption that Americans lack sufficient knowledge of the personal behaviors leading to weight gain. This is apparent in comparing the federal institution’s policies regarding obesity when compared to other disease states. For example, National Institute of Health’s (NIH) projected spending for 1 Epidemiology and Discrimination in Obesity HIV/AIDS in 2012 was $3.075 billion. When compared to this, obesity, which affects more individuals and poses numerous health risks, is allocated $830 million. The NIH is comprised of 27 institutes and centers, each with a specific research agenda, which focus on pressing and significant health issues. Some of these institutes include the National Cancer Institute, Eye Institute, the Human Genome Research Institute, the Institute of Allergy and Infectious Disease, and Institute of Mental Health to name a few. However, there is not an institute dedicated to obesity, arguably one of the most serious chronic diseases on both the national and international level. Federal and state legislative initiatives related to obesity have failed to address societal and environmental contributors for obesity. There is also a significant lack of attention to stigma associated with obesity and its consequences for individuals in public health efforts against this epidemic. Stigmatization of obese individuals poses serious risks to their psychological and physical health, generates health disparities, and interferes with implementation of effective obesity prevention efforts. To optimize obesity prevention and intervention efforts, these assumptions must be addressed within the sphere of public health, with recognition of the harmful impact of weight stigma on quality of life and the need to eliminate stigma from current and future public health approaches to the obesity epidemic. Experimental research has shown that providing individuals with information emphasizing personal responsibility for obesity increases negative stereotypes toward obese persons, whereas information highlighting the complex etiology of obesity (such as biological and genetic contributors) improves attitudes and reduces stereotypes [31]. Conclusion There is a clear need for increased public awareness and education about the complex etiology of obesity and the significant obstacles present in efforts to achieve sustainable weight loss. The prevailing societal and media messages that reinforce blame on obese persons need to be replaced with messages that obesity is a chronic disease with a complex etiology and is a lifelong condition for most obese persons. Supporting individuals with adaptive ways to cope with weight stigma can facilitate weight loss outcomes. Question Section 1. Overweight is defined as a BMI (kg/m2) of _______. A. 20–24.9 B. 30–34.9 C. >35 D. 25–29.9 13 2. Which of the following is true? A. Medical costs for obese individuals are the same as individuals with normal weight. B. Patients with “apple” distribution are considered to have a lower health risk than “pear” distribution. C. Women have more adiposity for the same BMI group. D. Adiposity decreases with age. 3. Which of the following races/ethnicities has the highest prevalence of obesity? A. Non-Hispanic White B. Non-Hispanic Asian C. Hispanic D. Non-Hispanic Black 4. Discrimination related to obesity has been documented in which of the following spheres? A. Employment B. Health-care facilities C. Educational institutions D. Close interpersonal relationships E. All of the above References 1. World Health Organization. http:// http://www.who.int/news-room/ fact-sheets/detail/obesity-and-overweight. Accessed 01 June 2018. 2. Hales CM, Carroll MD, Fryar CD, Ogden CL. Prevalence of obesity among adults and youth: United States, 2015–2016. HCHS Data Brief. No. 288. October 2017. 3. GBD 2015 Obesity Collaborators. Health effects of overweight and obesity in 195 countries over 25 years. N Engl J Med. 2017;377(1):13–27. 4. Crawley J, Meyerhoefer C. The medical care costs of obesity: an instrumental variables approach. J Health Econ. 2012;31(1): 219–30. 5. Bergman RN, Stefanovski D, Buchanan TA, Sumner AE, Reynolds JC, Sebring NG, et al. A better index of body adiposity. Obesity (Silver Spring). 2011;19(5):1083–9. 6. [No authors listed] Clinical guidelines on the identification, evaluation, and treatment of overweight and obesity in adults: executive summary. Expert panel on the identification, evaluation, and treatment of overweight in adults. Am J Clin Nutr. 1998;68:899–917. 7. http://whqlibdoc.who.int/hq/1998/WHO_NUT_NCD_98.1_(p1158).pdf. Accessed 15 May 2018. 8. Tchernof A, Després JP. Pathophysiology of human visceral obesity: an update. Physiol Rev. 2013;93(1):359–404. 9. http://www.cdc.gov/growthcharts/clinical_charts.htm. Accessed 28 May 2018. 10. World Health Organization. http://www.who.int/gho/ncd/risk_factors/obesity_text/en/. Accessed 01 June 2018. 11. Kelly T, Yang W, Chen CS, Reynolds K, He J. Global burden of obesity in 2005 and projections to 2030. Int J Obes. 2008;32(9): 1431–7. 12. Ng M, Fleming T, Robinson M, et al. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980–2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet. 2014;384(9945):766–81. 13. Ogden CL, Carroll MD. Prevalence of overweight, obesity, and extreme obesity among adults: United States, trends 1960–1962 through 2007–2008. National Center for Health Statistics. http:// 14 www.cdc.gov/nchs/data/hestat/obesity_adult_07_08/obesity_ adult_07_08.pdf. Accessed 21 May 2013. 14. Ogden CL, Lamb MM, Carroll MD, Flegal KM. Obesity and socioeconomic status in adults: United States, 2005–2008. NCHS Data Brief. 2010;50:1–8. 15. Ogden CL, Lamb MM, Carroll MD, Flegal KM. Obesity and socioeconomic status in children and adolescents: United States, 2005– 2008. NCHS Data Brief. 2010;51:1–8. 16. Fryar C, Carroll MD, Ogden CL. Prevalence of overweight, obesity, and extreme obesity among adults: United States, trends 1960–1962 through 2009–2010. NCHS. http://www.cdc.gov/nchs/data/hestat/ obesity_adult_09_10/obesity_adult_09_10.htm. Accessed 28 May 2013. 17. Fryar C, Carroll MD, Ogden CL. Prevalence of obesity among children and adolescents: United States, trends 1963–1965 through 2009–2010. NCHS. http://www.cdc.gov/nchs/data/hestat/obesity_ child_09_10/obesity_child_09_10.htm. Accessed 28 May 2013. 18. Ogden CL, Carroll MD, Kit BK, Flegal KM. Prevalence of obesity in the United States, 2009–2010. NCHS Data Brief. 2012;82:1–8. 19. US Department of Health and Human Services. http://www. healthypeople.gov/2020/topicsobjectives2020/objectiveslist. aspx?topicId=29. Accessed 28 May 2013. 20. Sturm R. Increases in morbid obesity in the USA: 2000–2005. Public Health. 2007;121(7):492–6. 21. NCD Risk Factor Collaboration (NCD-RisC). Worldwide trends in body-mass index, underweight, overweight, and obesity from 1975 to 2016: a pooled analysis of 2416 population-based measurement studies in 128·9 million children, adolescents, and adults. Lancet. 2017;390(10113):2627–42. 22. https://www.cdc.gov/obesity/data/childhood.html. Accessed 01 July 2018. 23. Field AE, Cook NR, Gillman MW. Weight status in childhood as a predictor of becoming overweight or hypertensive in early adulthood. Obes Res. 2005;13(1):163–9. 24. Greenberg JA. Obesity and early mortality in the United States. Obesity (Silver Spring). 2013;21(2):405–12. 25. Catenacci VAVA, Hill JO, Wyatt HR. The obesity epidemic. Clin Chest Med. 2009;30(3):415–44. 26. Mitchell NS, Catenacci VA, Wyatt HR, Hill JO. Obesity: overview of an epidemic. Psychiatr Clin North Am. 2011;34(4):717–32. 27. Ogden CL, Fakhouri TH, Carroll MD, et al. Prevalence of obesity among adults, by household income and education – United States, 2011–2014. MMWR Morb Mortal Wkly Rep. 2017;66:1369–73. 28. Hill JO, Wyatt HR, Reed GW, Peters JC. Obesity and the environment: where do we go from here? Science. 2003;299:853–5. 29. CDC. Facts about physical activity. https://www.cdc.gov/physicalactivity/data/facts.htm. Accessed 23 July 2018. R. A. Forse et al. 30. Andreyeva T, Puhl RM, Brownell KD. Changes in perceived weight discrimination among Americans, 1995–1996 through 2004–2006. Obesity (Silver Spring). 2008;16(5):1129–34. 31. Puhl RM, Heuer CA. Obesity stigma: important consideration for public health. Am J Public Health. 2010;100(6):1019–28. 32. Roehling MV, Roehling PV, Pichler S. The relationship between body weight and perceived weight-related employment discrimination: the role of sex and race. J Vocat Behav. 2007;71(2):300–18. 33. Budd GM, Mariotti M, Graff D, Falkenstein K. Health care professionals’ attitudes about obesity: an integrative review. Appl Nurs Res. 2011;24(3):127–37. 34. Baicker K, Cutler D, Song Z. Workplace wellness programs can generate savings. Health Aff (Millwood). 2010;29(2):304–11. 35. Finkelstein E, Trogdon J, Cohen J, Dietz W. Annual medical spending attributable to obesity: payer- and service-specific estimates. Health Aff. 2009;28(5):w822–31. 36. Pantenburg B, Sikorski C, Luppa M, et al. Medical students’ attitudes towards overweight and obesity. Pereira RI, ed. PLoS ONE. 2012;7(11):e48113. 37. Amy NK, Aalborg A, Lyons P, Keranen L. Barriers to routine gynecological cancer screening for White and African-American obese women. Int J Obes. 2006;30(1):147–55. 38. Barofsky I, Fontaine KR, Cheskin LJ. Pain in the obese: impact on health-related quality-of-life. Ann Behav Med. 1997;19(4):408–10. 39. Jackson SE, Beeken RJ, Wardle J. Obesity, perceived weight discrimination and psychological well-being in older adults in England. Obesity (Silver Springs) 2015; 23(5):1105–11. 40. Turnbull JD, Heaslip S, McLeod HA. Pre-school children’s attitudes to fat and normal male and female stimulus figures. Int J Obes. 2000;24:1705–6. 41. Puhl RM, King KM. Weight discrimination and bullying. Best Pract Res Clin Endocrinol Metab. 2013;27(2):117–27. 42. Griffiths LJ, Wolke D, Page AS, Horwood JP. Obesity and bullying: different effects for boys and girls. Arch Dis Child. 2006;91(2):121– 5. https://doi.org/10.1136/adc.2005.072314. 43. Pasquale RD, Celsi L. Stigmatization of overweight and obese peers among children. Front Psychol. 2017;8:524. 44. Cramer P, Steinwert T. Thin is good, fat is bad: how early does it begin? J Appl Dev Psychol. 1998;19:429–51. 45. Kraha A, Boals A. Parents and vehicle purchases for their children: a surprising source of weight bias. Obesity (Silver Spring). 2011;19(3):541–5. 46. Crandall C. Do parents discriminate against their heavyweight daughters? Personal Soc Psychol Bull. 1995;21:724–35. 47. Gortmaker SL, Must A, Perrin JM, Sobol AM, Dietz WH. Social and economic consequences of overweight in adolescence and young adulthood. N Engl J Med. 1993;329:1008–12. 2 The Pathophysiology of Obesity and Obesity-Related Disease Robert W. O’Rourke Abbreviations AgRP ARCN ATM BAT BMI CART CNS DIT ER GIP GLP-1 GWAS HFC LCFA LHA MAPK MCH MSH NAFLD NEAT NO NPY NREE POMC PUFA PVN REE ROS SAT SNP SVF Agouti-related peptide Arcuate nucleus Adipose tissue macrophages Brown adipose tissue Body mass index Cocaine- and amphetamine-regulated transcript Central nervous system Diet-induced thermogenesis Endoplasmic reticulum Glucose-dependent insulinotropic polypeptide Glucagon-like peptide-1 Genome-wide association analysis Hypothalamic feeding center Long-chain fatty acids Lateral hypothalamic area Mitogen-activated protein kinases Melanin-concentrating hormone Melanocyte stimulating hormone Nonalcoholic fatty liver disease Non-exercise-induced thermogenesis Nitric oxide Neuropeptide Y Non-resting energy expenditure Pro-opiomelanocortin Polyunsaturated fatty acids Paraventricular nucleus Resting energy expenditure Reactive oxygen species Subcutaneous adipose tissue Single nucleotide polymorphism Stromal-vascular cell fraction R. W. O’Rourke (*) Department of Surgery, University of Michigan, Ann Arbor, MI, USA Bariatric Surgery Program, Division of General Surgery, Ann Arbor Veterans Administration Hospital, Ann Arbor, MI, USA e-mail: rorourke@med.umich.edu TEE UCP UPR VAT WAT Total energy expenditure Uncoupling proteins Unfolded protein response Visceral adipose tissue White adipose tissue Chapter Objectives • Part I 1. Understand the mechanisms that regulate body weight and their contribution to the obesity phenotype, including regulation of satiety and hunger, metabolic rate, and thermogenesis. 2. Understand the evidence that demonstrates that obesity is a genetic phenomenon, including data from twin studies and genome-wide analyses. 3. Understand the concept of thrifty genes, the thrifty genotype, epigenetics, and the evolutionary influences that contribute to the human tendency towards excess adiposity. 4. Understand the terms homeostasis, allostasis, and homeorhesis and how these terms describe the behavior of biologic systems. • Part II 1. Understand the early events in adipose tissue that contribute to adipose tissue dysfunction including hypoxia and nutrient excess. 2. Understand how endoplasmic reticulum stress, oxidative stress, and inflammation evolve in adipose and other tissues and how these phenomena conspire to induce metabolic dysfunction at the tissue level. 3. Understand the concepts of adipose tissue buffering and overflow and their contribution to the pathogenesis of metabolic disease. 4. Understand the basic concepts behind the pathogenesis of liver disease, vascular disease, diabetes, and cancer in obesity. © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_2 15 16 R. W. O’Rourke Introduction Obese people… already subject to adverse health effects are additionally victimized by a social stigma predicated on the Hippocratic nostrum that weight can be controlled by ‘deciding’ to eat less and exercise more. This simplistic notion is at odds with substantial scientific evidence illuminating a precise and powerful biologic system that maintains body weight within a relatively narrow range. Voluntary efforts to reduce weight are resisted by potent compensatory biologic responses. -Jeffrey Freidman [1] The current epidemic of obesity is partly caused by the fact that we all possess an ancient metabolism selected to protect us from starvation, and hence, quite unsuited to our modern lifestyle…the metabolisms of most of humankind have been honed by famine and starvation...hunger and famine have been an everpresent influence on genetic selection. -Andrew Prentice [2] Obesity has been a part of the human condition since our genesis. Stone figurines depicting obesity found throughout Europe are among the earliest human artifacts [3] (Fig. 2.1). Obesity is not a new phenomenon but rather part and parcel of the human condition, and in our modern environment, the majority of the population is overweight or obese [4]. Lifestyle modification achieves significant durable weight a loss only rarely. Why is this failure rate so high? Friedman alludes to powerful biologic systems that defend body weight, systems rooted in the midbrain and molded by a genetic heritage that was forged, as Prentice states, by the selective pressure of famine present during our evolution. An understanding of the pathophysiology of obesity and metabolic disease will guide development of novel therapeutic interventions. Perhaps equally important, an understanding of the origins of the epidemic and an appreciation that obesity in most cases results not from a defect of individual willpower but rather from a physiology shaped by eons of genetic selection and thrust into a modern “obesogenic” environment provide a basis for empathy towards those afflicted with a debilitating condition. This chapter reviews the physiologic mechanisms that lead to the obesity phenotype, answering the question “How do we become obese?” We go on to discuss genetic, evolutionary, and environmental forces that molded these regulatory systems to create the modern epidemic, answering the question “Why do we become obese?” If obesity was simply a cosmetic condition, we might end there, but obesity is associated with a wide range of pathology. We will explore the b Fig. 2.1 (a) Mammoth ivory figurine, Swabian Jura, Germany circa 35,000 B.C. (b) Venus of Willendorf, circa 22,000 B.C. 2 The Pathophysiology of Obesity and Obesity-Related Disease 17 pathophysiology of metabolic disease and the effects of nutrient excess on cellular metabolism and systemic physiology. Pathophysiology of Obesity Pathophysiologic Mechanisms ow Do We Become Obese? H Why do diets fail? Why do dieters hit a “wall” that resists further weight loss? Why, in short, can we not simply “decide” to eat less? The answers to these questions are rooted in the physiologic systems that regulate body weight. Humans are creatures of behavior, and so the most important of these mechanisms control food-related behavior. But the processes that regulate energy homeostasis are so central to our biology that they interface with every aspect of physiology. These processes include control of food intake, energy expenditure, thermogenesis, adipocyte biology, and a host of other metabolic processes. ontrol of Food Intake: The Leptin Paradigm C The reason we become obese, at the most basic level, is that we eat more calories than we metabolize. The most important weight regulatory mechanism in humans is the collective system that controls food intake. Hunger, defined as the desire to eat, and satiety, defined as the lack of hunger, describe fundamental aspects of eating behavior. While conceptually useful, at the cellular and molecular level, these definitions are less precise, as satiety and hunger mediators utilize distinct yet intimately associated signaling pathways. Furthermore, the dichotomy between satiety and hunger is overly simplistic, as feeding behavior is highly complex and includes subtle behaviors such as eating speed, food preferences, hunger irritability, and sensory and emotional responses to food. We often consider such behaviors to be under conscious control, but in fact the neural networks that control these behaviors reside in the midbrain, an area that from an evolutionary perspective long predates the frontal cortex, the seat of conscious thought and cognition. Eating behavior is regulated predominantly at a subconscious level. We can, for a time, consciously control our food intake and lose weight with dieting, but with few exceptions, such efforts are limited in magnitude and followed by weight regain, as midbrain counter-regulatory systems are activated that vigorously defend body weight. In 1994 Dr. Jeffrey Friedman cloned the leptin gene [5]. Friedman studied the Ob mouse, a hyperphagic obese strain described in the 1950s that lacked a circulating factor that, when restored from wild-type mice in parabiosis experiments, reversed the obese phenotype (Fig. 2.2). Friedman identified circulating factor as the 16kD protein leptin and Fig. 2.2 The Ob mouse found the leptin gene to be mutated and nonfunctional in the Ob mouse. Restoration of exogenous wild-type leptin protein to Ob mice reversed obesity. Friedman’s group cloned the human leptin gene, and within a few years, two obese human kindreds were identified with similar leptin mutations in whom administration of exogenous leptin reversed obesity [6]. These results were met with much excitement, and leptin was considered a potential treatment for obesity in the general population. It was soon found, however, that single monogenic leptin mutations are a rare cause of common human obesity. Exogenous leptin, while a cure for Ob mice and Ob humans, had no therapeutic effect in most obese humans. In fact, common human obesity is characterized by elevated leptin levels and resistance to leptin’s satiety effects, a phenomenon not dissimilar from insulin resistance [7, 8]. The discovery of leptin transformed our understanding of energy metabolism. Leptin mediates a complex communication between the gut, adipose tissue, and brain that regulates food intake. Secreted by adipose tissue in response to a meal, leptin circulates through the bloodstream to bind its receptor in the hypothalamus and effect satiety. Serum leptin levels are low after a nighttime fast; breakfast generates an adipose tissue leptin secretory response that induces satiety and limits food intake for a few hours, after which leptin levels wane and we find ourselves hungry by lunch and the cycle repeats. In this manner, leptin controls short-term food intake. Leptin also regulates long-term food intake and adipose tissue stores. Peak postprandial leptin levels are determined by the adipose tissue mass available to secrete leptin, which in turn dictates the magnitude and kinetics of the satiety response. When adipose tissue mass is reduced with dieting, postprandial leptin levels decrease, chronically attenuating the postprandial satiety response, leading to a progressive increase in food intake until adipose tissue mass and postprandial leptin levels are restored to baseline levels. This mechanism provides a cogent explanation for weight regain after dieting. he Hypothalamic Feeding Center T As early as 1940, hypothalamic ablation experiments in rats that induced hyperphagia and obesity first demonstrated the 18 R. W. O’Rourke central role of the hypothalamus in weight regulation. Leptin tem (CNS) regions, as well as remote organs including adibinds receptors within the arcuate nucleus (ARCN) of the pose tissue and liver, that relay information regarding hypothalamic feeding center (HFC) and activates anorexi- short-term prandial status and long-term energy stores. In genic (satiety-inducing), catabolic pro-opiomelanocortin response, the HFC orchestrates diverse behavioral, meta(POMC), and cocaine- and amphetamine-regulated tran- bolic, and physiologic responses via efferent projections to script (CART)-producing first-order neurons in the dorsolat- multiple brainstem and higher level central nervous system eral ARCN. POMC is posttranslationally processed to yield networks that regulate food intake. These communication the anorexigenic peptides α-, β-, and γ-MSH. Leptin simul- networks overlap with CNS regions involved in emotional taneously inhibits secretion of the orexigenic (hunger-­ responses, cognition, sensory processing, and memory. Food inducing) mediators neuropeptide Y (NPY) and agouti-related intake activates reward centers, hedonic circuits that drive peptide (AgRP) by the ventromedial ARCN. First-order consumption of foods high in fat, sugar, and salt in the ARCN neurons in turn project to second-order neurons, absence of caloric deficiency; visual food stimuli activate including those within the paraventricular nucleus (PVN) dedicated memory and visual circuits linked to emotional and lateral hypothalamic area (LHA), which in turn elabo- responses [10]. Food acquisition is such an important task in rate secondary signaling mediators. The PVN generates an our evolutionary history that it involves all areas of the brain anorexigenic, catabolic program via expression of CRH, and all aspects of behavior and physiology. TSH, and oxytocin, which increase satiety and energy expenditure via multiple endocrine and autonomic nervous system A Complicated Family of Mediators pathways. The LHA, in contrast, generates an orexigenic, Leptin opened the door to the discovery of a family of proanabolic program via expression of orexins A and B and teins that regulate food intake, broadly classified as adipomelanin-concentrating hormone (MCH). Each of these path- kines, gut hormones, and cytokines (Table 2.1). Adipokines ways inhibits the other at the level of first-order neuron trans- are secreted predominantly by adipose tissue, gut hormones mitter effects on second-order neurons [9] (Fig. 2.3). by the gastrointestinal tract and viscera, and cytokines by The HFC exhibits an intrinsic set point that vigorously leukocytes. These proteins share phylogenetic and functional defends adipose tissue stores, the so-called adipostat. The overlap and classification systems remain in flux. For examHFC is influenced by multiple afferent inputs from all organ ple, leptin is a member of the long-chain helical chain cytosystems, including diverse neighboring central nervous sys- kine family, of which IL-6 and IL-11 are members; the term adipocytokines is sometimes used to reflect the close relationship between adipokines and cytokines. In many cases Orexigenic, Anorexigenic, these mediators are secreted by multiple tissues and cell anabolic signals catabolic signals types. TNF-α is expressed predominantly by macrophages but also by adipocytes; ghrelin is secreted primarily by gastric fundus cells but is also expressed at low levels by the MCH, orexins A,B CRH, TRH, oxytocin placenta, kidney, pituitary, endometrium, macrophages, and hypothalamus; adipocytes are the dominant source of leptin, LHA Second order neurons PVN but small amounts are also secreted by the placenta, muscle, and stomach. + + Most but not all adipokines, gut hormones, and cytokines NPY, AgRP POMC, CART, α -MSH regulate food intake. Leptin appears to play a dominant role in regulating long-term satiety, while the gut hormones CCK, Dorsolateral Ventromedial PYY, and ghrelin dominate short-term food intake including First order neurons ARCN ARCN variables such as meal size and duration [9]. Ghrelin is + secreted during fasting and binds receptors in the hypothalamus to induce an orexigenic signal. Other mediators have Leptin only modest effects on food intake. Adiponectin, an adipokine secreted by adipocytes, stimulates insulin secretion, Fig. 2.3 A simplified schematic of the hypothalamic feeding center: inhibits pancreatic beta-cell apoptosis, and attenuates inflamFirst-order ARCN neurons communicate with second-order PVN and LHA neurons to coordinate behavioral and metabolic output. PVN sig- mation. Consistent with these beneficial effects, serum adinaling is primarily anorexigenic and catabolic and enhanced by leptin ponectin levels are inversely correlated with obesity and and insulin, while LHA signaling is primarily orexigenic and anabolic metabolic disease. and inhibited by leptin and insulin. Both pathways negatively regulate In addition to regulating food intake, satiety and hunger the other. Other peripheral and central mediators (not shown) stimulate mediators manifest multiple other functions. Leptin induces first- and second-order neurons, including insulin, ghrelin, CCK, GLP-­ 1, serotonin, endogenous cannabinoids, and norepinephrine macrophage, monocyte, and T-cell proliferation and cytokine 2 The Pathophysiology of Obesity and Obesity-Related Disease 19 Table 2.1 Proteins involved in weight regulation Mediator Adipokines Adiponectin Apelin Primary source Satiety Glucose homeostasis Immunity Minimal satiety effects Probably anorexigenic, data sparse Unknown Leptin Adipocytes Pro-inflammatory, macrophage homing Generally pro-inflammatory Lipocalin 2 Adipocytes, monocytes, macrophages Adipocytes Anorexigenic, hypothalamic leptin resistance in obesity Unknown Insulinomimetic Inhibits glucose-induced insulin secretion Diabetogenic, likely through pro-inflammatory properties Generally insulinomimetic Anti-inflammatory Anti-inflammatory CCL2 Adipocytes Adipocytes, brain, heart, kidney, lung Adipocytes Diabetogenic via pro-­ inflammatory effects Pro- and anti-inflammatory effects Unknown Unknown Probably diabetogenic – causality not well-established Diabetogenic via pro-­ inflammatory effects Diabetogenic Unknown Insulinomimetic Anti-inflammatory, suppresses Wnt signaling Plasminogen activator inhibitor-1 (PAI-1) Resistin Retinol-binding protein 4 (RBP-4) Adipocytes, macrophages Adipocytes, hepatocytes, macrophages Adipocytes, pancreas Unknown Probably anorexigenic Pro-inflammatory Pro-inflammatory Secreted frizzled-­ related protein 5 (SFRP5) Visfatin Adipocytes May induce satiety, data conflicting Insulinomimetic Pro-inflammatory Gut hormones Amylin Pancreatic β cells Anorexigenic, increases energy expenditure Probably pro-inflammatory, conflicting data Duodenal, jejunal I cells, CNS Gastric fundal cells Duodenal, jejunal C cells Anorexigenic, regulates meal termination Orexigenic Anorexigenic Co-released with insulin in response to food, inhibits insulin secretion Probably insulinomimetic Anti-inflammatory Diabetogenic Induces insulin secretion, conflicting data Anti-inflammatory Pro- and anti-inflammatory effects, conflicting data Cholecystokinin (CCK) Ghrelin Glucose-dependent insulinotropic peptide (GIP) Glucagon-like peptide-1 (GLP-1) Glucagon Ileal L cells, CNS Anorexigenic Insulinomimetic Anti-inflammatory Pancreatic α cells Pancreatic β cells Increases blood glucose levels, insulin secretion “Insulinomimetic” Probably pro-inflammatory Insulin Anorexigenic, increases energy expenditure Anorexigenic, increases energy expenditure Oxyntomodulin Ileal L cells Probably insulinomimetic Pancreatic polypeptide (PP) Peptide tyrosine tyrosine (PYY) Cytokines IFN-γ Pancreatic F cells Ileal L cells Anorexigenic, increases energy expenditure Anorexigenic, increases energy expenditure Anorexigenic Insulinomimetic, increases hepatic insulin sensitivity Insulinomimetic T cells, NK cells Unknown Generally diabetogenic Macrophages Adipocytes, macrophages, lymphocytes Macrophages, lymphocytes Adipocytes, macrophages, lymphocytes Anorexigenic Unknown, likely anorexigenic in context of cachexia Unknown Diabetogenic Generally diabetogenic although effects are context dependent and conflicting Insulinomimetic Anti-inflammatory Anorexigenic, mediates cachexia response Diabetogenic Pro-inflammatory IL-1 IL-6 IL-10 TNF-α Generally anti-inflammatory, CNS administration may promote inflammation Unknown Unknown Anti-inflammatory, inhibits NFκB activation Pro-inflammatory, induces macrophage inflammation Pro-inflammatory Generally pro-inflammatory 20 expression, with a bias towards generating pro-­inflammatory responses [11]. Leptin controls endocrine function, potentiating pituitary-hypothalamic axis activity and regulating steroid metabolism. Leptin is implicated in cognitive function, learning, and memory; leptin-deficient humans suffer mild cognitive deficits that are corrected with exogenous leptin replacement [12]. The incretins are a class of gut hormones that includes glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-­1). GIP is secreted by K cells in the duodenum and jejunum in response to a glucose load, induces insulin secretion by pancreatic β cells, and stimulates fatty acid synthesis in adipocytes. GLP-1 is secreted by L cells in the ileum and has insulinomimetic properties, decreasing peripheral insulin resistance and potentiating pancreatic insulin secretion. GLP-1 also inhibits gastric emptying and induces satiety [13]. Altered secretion of incretins secondary to anatomic changes in the intestinal tract has been implicated in metabolic responses to bariatric surgery, although exact mechanisms remain unclear. Furthermore, GLP-1, GIP, and other gut hormones have immunoregulatory functions. Similar functional diversity characterizes adipocytokines. TNF-α induces insulin resistance and altered lipid metabolism and promotes anorexia in cachexia and inflammatory states [14]. In general, adipose tissue cytokines and adipokines are either pro-inflammatory and diabetogenic (e.g., TNF-α, IL-6, IL-8, IL-1, resistin) or anti-inflammatory and insulinomimetic (e.g., IL-10, IL-1Ra, adiponectin), with the former upregulated and the latter downregulated in obesity. Exceptions exist: the orexigen ghrelin, for example, has anti-­inflammatory properties but induces insulin resistance [15]. Multiple other mediators regulate feeding behavior, including insulin, endogenous opioids, adrenergic agonists, and cannabinoids. Redundancy, complex regulatory control, and functional pleiotropy involving virtually all physiologic systems characterize the broad family of molecules that regulate food intake. Energy Expenditure While dominant, control of food intake is not the sole mechanism of weight regulation. Obese patients often report eating little but suffering from a “slow metabolism.” The magnitude of the effect of differences in energy expenditure on human body weight variability is debated but, while less than contributions from differences in the regulation of food intake, nonetheless contributes. Metabolic rate has been studied in response to weight loss in healthy volunteers using indirect calorimetry. In conditions of energy deficit, total energy expenditure (TEE), comprised of resting energy expenditure (REE, 60–70% of TEE, defined as energy used for basic physiologic functions at rest) and non-resting energy expenditure (NREE, 30–40% of TEE, defined as energy used for activity above and beyond REE), is decreased R. W. O’Rourke beyond that expected by loss of fat and fat-free mass alone. These changes are secondary to equal decreases in NREE and REE and accompanied by a corresponding decrease in voluntary physical activity. Similar changes are observed in obese subjects who achieve 10–20% body weight loss, whose TEE decreases up to 20% more relative to predicted values from loss of fat and fat-free mass [16, 17]. Obese subjects who maintain weight loss have lower resting metabolic rates than lean subjects, changes that persist for years, and mandate constant dietary vigilance [18]. In contrast to diet-­ induced weight loss, bariatric surgery-induced weight loss may be paradoxically associated with increased TEE in rodents and humans. In the absence of surgery, however, compensatory decreases in energy expenditure counteract caloric restriction and provide yet another explanation for the failure of dietary weight loss efforts. Studies of overfeeding and weight gain in obese subjects demonstrate converse changes with notable differences. First, overfeeding leads to a compensatory increase in TEE primarily due to increased NREE, in contrast to weight loss, in which equal decreases in NREE and REE are observed [16, 19]. Second, voluntary overfeeding studies in twin cohorts demonstrate a significant genetic contribution to individual variability in weight gain in response to overfeeding [20]. Third and importantly, increases in TEE in response to overfeeding appear to be transient, lasting months then reverting to baseline, unlike sustained weight loss for which such changes tend to be long-lasting. Furthermore, while compensatory increases in TEE oppose weight gain, this effect is weaker than that of decreased TEE with weight loss. Taken together, these observations suggest that regulation of metabolic rate is more robust in conditions of negative rather than positive energy balance – the system is better designed to prevent leanness than avoid adiposity, a common theme. The mechanisms underlying the regulation of energy expenditure and metabolic rate are complex, but sympathetic nervous and hormonal systems are dominant. Voluntary choice controls NREE to some extent, but much like food intake, we consciously control our activity levels to a lesser degree than we may imagine. Differences in autonomic nervous system activity contribute to variability in metabolic rate: overfeeding increases and underfeeding decreases sympathetic nervous system activity in rodents, and variability in sympathetic nervous system activity is observed in humans, with higher levels predicting successful weight loss [21]. Obese subjects manifest alterations in thyroid hormone and catecholamine balance that are associated with decreased metabolic rates [22]. At the cellular level, these variables control energy expenditure by regulating thermogenesis. Thermogenesis In 1783 Laplace and Lavoisier invented the first calorimeter and measured energy balance in animals, demonstrating that 2 The Pathophysiology of Obesity and Obesity-Related Disease biologic systems obey the first law of thermodynamics, such that energy intake equals the sum of energy storage and energy expenditure. Energy expenditure in turn may be approximated by the sum of energy used to drive physical activity and cellular processes, and energy dissipated as heat. As Lavoisier famously concluded, “Life is combustion.” All cellular biochemical reactions are less than 100% efficient and thus generate heat. Thermogenesis occurs in all cells but is most active in brown adipose tissue (BAT) and skeletal muscle. Skeletal muscle thermogenesis occurs during exercise, during movement not associated with exercise (non-­ exercise-­ induced thermogenesis, NEAT), and during cold-induced shivering. Skeletal muscle energy utilization efficiency during exercise is increased in subjects who lose weight and decreased in subjects who gain weight [17]. Furthermore, lower levels of NEAT have been demonstrated in obese humans [23], suggesting that differences in skeletal muscle thermogenesis contribute to the pathogenesis of obesity. BAT is a dominant site of thermogenesis, in which it underlies non-shivering thermogenesis, a critical thermoregulatory mechanism in rodents and neonatal humans. BAT, relative to white adipose tissue (WAT), is associated with higher thermogenic potential, increased mitochondrial number and function, decreased lipid storage, and improved metabolism [24]. Diet-induced thermogenesis (DIT), a postprandial increase in metabolic rate observed in rodents and humans, is an important component of the thermogenic response. At the cellular level, DIT increases mitochondrial heat production and decreases energy extraction, limiting exposure to postprandial increases in nutrient flux. Humans demonstrate significant variability in the magnitude of DIT, with obese humans having lower DIT responses [25]. While all biochemical reactions generate heat, membrane ion leaks that occur in all cells and triglyceride-fatty acid futile cycling within skeletal muscle are examples of processes that involve significant thermogenesis. Uncoupling of electron transport from oxidative phosphorylation within mitochondria is a dominant-regulated thermogenic process. Mitochondria, evolved from intracellular parasites to become endosymbiotic cell organelles, utilize oxidative phosphorylation coupled to electron transport along their inner membrane to generate ATP from glycolytic energy substrates. The efficiency of this process is regulated by uncoupling proteins (UCPs) that uncouple oxidative phosphorylation from electron transport, creating a proton leak that effectively runs the mitochondrial engine in neutral, generating heat rather than ATP. UCPs adjust mitochondrial efficiency and act as a thermoregulatory mechanism. This control mechanism extends across animal and plant phyla: uncoupling occurs in potatoes in response to cold weather [19]. Three dominant human UCP isoforms are identified. UCP-1 expression is restricted to BAT; UCP-2 is widely expressed in multiple tissues, while UCP-3 is expressed in the BAT, skeletal muscle, and brain. 21 In humans the correlation between UCP function and thermogenesis specifically, and metabolic rate in general, is imperfect. Skeletal muscle UCP expression is paradoxically upregulated in response to starvation, for example, confusing the role of uncoupling in response to nutrient deficit [26]. The type and quantity dietary constituents, along with variables such as environmental temperature and photoperiod, also influence UCP-1 expression, adding to complexity. Finally, the magnitude, duration, and direction of changes in UCP-1 expression in BAT in response to overfeeding in rodents are heterogeneous and strain-dependent, reinforcing the importance of genetic background on the regulation of metabolic rate. Nonetheless, a transgenic mouse overexpressing UCP-1 under control of the adipose tissue-specific AP2 promoter is resistant to obesity [27], and polymorphisms in UCP genes correlate with obesity and metabolic disease in humans [28], supporting a role for UCPs in obesity pathogenesis. dipocyte Biology and Other Metabolic A Processes Differences in the kinetics of adipocyte hypertrophy, proliferation, differentiation, and metabolism contribute to variability in body weight. It was once thought that adipocytes did not proliferate or increase in number in adults, but recent data demonstrate otherwise. Obese subjects, especially those with early-onset childhood obesity, have a greater number of adipocytes than lean subjects, as well as preadipocyte precursors with a metabolically adverse phenotype and decreased proliferative potential [29]. The regulatory systems described above comprise only a partial list of the many physiologic processes that occur within adipose and other tissues that control energy balance, all of which are regulated by homeostatic systems with differing amplitudes and kinetics that contribute to variability in human body weight. Homeostasis Despite the power of weight regulatory systems, we have a degree of control over our body weight, and as such within individual limits, personal choice plays a role in the pathogenesis of obesity. But the magnitude of the effect of personal choice is less than we may believe, as food intake is subject to tightly regulated subconscious mechanisms. The limits imposed by these mechanisms are powerful and indeed represent a fundamental characteristic of all biologic systems, all of which have one goal: the maintenance of homeostasis. Perfect homeostasis is of course elusive: our heart rate is not a constant 72 beats per minute; rather we are constantly buffeted by external forces that drive us away from the homeostatic mean. In the case of energy homeostasis, this buffeting comes in the form of periodic alterations in food resources, physical activity, illness, temperature, and innumerable other environmental and physiologic variables. 22 R. W. O’Rourke The term allostasis, proposed by Sterling and Eyer in 1988, describes how biologic systems evolved to achieve stability not through strict adherence to a homeostatic mean but rather through a dynamic response to external stimuli, the allostatic load, that achieves controlled variability around a homeostatic mean within a defined allostatic range. The robustness of biologic systems derives from this capacity for dynamic allostasis. Biologic systems function well within their allostatic range, deviation from which may be lethal. Furthermore, most humans, most strains of laboratory rodents, and many wild animals gradually gain weight over the course of their lives, suggesting not only a predisposition to excess weight but also towards gradual lifelong increase. Biologist Conrad Hal Waddington proposed the term homeorhesis, in contrast to homeostasis, to describe regulation of a system along such a trajectory [30] (Fig. 2.4). Leptin represents a paradigm for allostatic behavior but is by no means unique; all aspects of metabolism, indeed all biologic processes, are governed by allostasis. When we ask obese patients to lose significant weight with diet and exercise, we are asking them to step outside their allostatic range, an impossible task from a biologic perspective. This is why efforts to address the obesity crisis at the individual level fall short. This is why diets fail. To impact on the obesity epidemic, we must understand the genetic, evolutionary, and environmental influences that define metabolic allostatic range and how these factors conspire so vigorously to defend body weight. Evolutionary and Genetic Contributors hy Do We Become Obese? W Rare humans suffer a paucity of adipose tissue secondary to congenital and acquired lipodystrophy syndromes. These patients have voracious appetites, require high caloric intake to maintain lean body weight, and suffer from diabetes and steatosis, stressing the central role of adipose tissue in metabolism and demonstrating that its absence is as detrimental as its excess. The explanation for why humans are prone to excess adiposity but stringently protected from underweight conditions is rooted in genetics and influenced by powerful environmental and evolutionary selective pressures. Despite knowing that it is unhealthy, humans, along with many animals not capable of such knowledge, will eat to excess if surplus food is available. Hedonic circuits, mentioned above, and metabolic thrift, discussed below, are examples of physiologic and evolutionary mechanisms underlying this phenomenon. But why did evolution select for such behavior if it is detrimental to health? The reason is that evolution did not mold us to optimize long-term health or longevity. Rather, evolution selects for physiology and behavior that optimizes reproductive fitness. Despite its detrimental long-term effects, surplus nutrition, to a point, optimizes reproductive fitness in youth, as evidenced by increased fertility rates and younger age of onset of puberty in Western societies [31]. Reproduction is energy-intensive Allostatic load Homeorhetlc trajectory Body weight, satiety, leptin levels, other mediator levels, all metabolic and biologic processes, and many of the stimuli that regulate them Homeorhetic mean Allostatic range Allostatic load Fig. 2.4 Homeostasis, allostasis, homeorhesis: Levels of leptin and other mediators, the outcome measures they regulate (food intake, thermogenesis, and all other metabolic and physiologic processes), as well as physical activity and many environmental and physiologic stimuli that regulate these processes, all cycle in an allostatic manner around a homeostatic mean, or alternatively in some cases, along a homeorhetic trajectory (e.g., body weight over the course of a lifetime). External stimuli (the allostatic load) cause deviation from the homeostatic mean. Optimal functioning of a biologic system occurs within its allostatic range, defined as the degree to which a biologic system can tolerate deviation from its homeostatic mean without disruption 2 The Pathophysiology of Obesity and Obesity-Related Disease and evolution selected for a propensity to maximize energy resources to ensure adequate reproductive potential. Humans, unlike other animals, have the ability to self-reflect and, to an extent, “decide” to eat less and thus possibly increase long-­ term health and longevity, but as discussed, this ability is constrained by allostatically governed mechanisms. The distinct and separate goals of reproductive fitness and long-term health and longevity, the former hardwired by evolution, the latter unique to human cognition, lead to our conflict with overeating. Metabolic Thrift James Neel at the University of Michigan proposed the thrifty genotype hypothesis in 1962 [32]. Neel postulated that throughout evolution, the constant pressure of famine led to selection of genes that regulated metabolism in a thrifty manner: polymorphisms in metabolic genes were selected for if they imparted a tendency towards efficient energy storage and metabolism, while polymorphisms that imparted a tendency towards a less efficient metabolism were strongly selected against. Metabolic thrift provided a robust reproductive advantage in a food-sparse leptogenic environment but leads to a blossoming of the obesity phenotype in an obesogenic environment in which food is plentiful, exactly what we observe today. Polymorphisms resulting from random Darwinian mutation that predisposed to overweight, obesity, and thrift accumulated in the human genome, while those predisposing to leanness and a lack of thrift became rare. The thrifty gene hypothesis has evolved and remains debated. Thrifty gene candidates are sparse and proof of causality remains elusive. Alternative hypotheses such as predation release and genetic drift may also contribute to obesity [33, 34]. Despite these controversies, the thrifty genotype hypothesis provides a coherent explanation for the human predisposition towards overweight and obesity and its dramatic recent increase. Why Adipose Tissue? Animals employ diverse strategies for metabolic thrift, including hibernation, high reproductive rates, and high caloric intake. Among these strategies, storage of calories for future use is common. Storage may be ectopic: hamsters store up to 50% of their body weight in their cheek pouches. Humans in contrast, along with many other species, store energy in the form of adipose tissue. Adipose tissue is present across phyla, including yeast and insects [29], but is particularly well-developed in vertebrates. Adipose tissue is particularly energy dense – humans can store less than 1 day’s worth of calories in skeletal muscle in the form of glycogen but over 2 month’s worth of calories in fat in adipose tissue. Humans invest significant resources into a single offspring with each pregnancy, and human development is characterized by a prolonged period of rapid postnatal 23 growth and brain development that requires a constant supply of calories. Nutritional deficits during this period have widespread detrimental effects and exerted a strong reproductive disadvantage during evolution. Adipose tissue provides a constant source of calories during this critical period, reflected by the fact that human percent body fat is highest during this rapid neonatal growth phase. Adipose tissue thus protects our fragile, singleton, energy-hungry offspring. Finally, humans are to some extent a migratory species: much of our evolution was shaped by the out-of-Africa emigration that led to pan-global colonization 50,000– 75,000 years ago [35]. Adipose tissue allowed us to weather variability in food supplies during famines and migrations. For all these reasons, adipose tissue is a particularly good strategy for metabolic thrift in humans. he Genetic Basis of Obesity T Obesity has a strong genetic basis. In addition to leptin mutations, over 20 rare inborn genetic syndromes are associated with human obesity. In virtually all, obesity is one of a number of phenotypic abnormalities, emphasizing the interaction of weight regulation with all aspects of physiology. Causative genes have yet to be identified for Prader-Willi, Bardet-­ Beidl, Cohen, and Alstrom syndromes, while in other syndromes, specific genes have been implicated, many associated with feeding behavior, stressing the dominance of these systems in obesity pathogenesis. Some disorders are monogenic and include mutations in genes encoding POMC, PCSK1, and MC4R. But in contrast to monogenic obesity syndromes and similar to many chronic diseases, common human obesity is a complex polygenic phenotype, significantly complicating analysis. Despite these challenges, statistical genetics methods have accurately quantified the cumulative genetic contribution to obesity and demonstrate a high level of hereditability. Quantitative genetic analysis of twin cohorts is a powerful tool that is widely used to quantify genetic contribution to complex polygenic diseases and clinical phenotypes. Twin studies rely on comparison of thousands of monozygotic and dizygotic twin pairs to quantify the contribution of genetic influences to phenotypic traits based on Mendelian genetic inheritance with a high degree of analytic power. Further detail can be gleaned by study of twin pairs separated at birth. When applied to obesity, multiple studies consistently demonstrate that genetic factors dictate approximately 70% of the tendency towards a particular body habitus phenotype, while nongenetic, presumably environmental factors contribute approximately 30% [20]. Twin studies also reveal enlightening aspects of food-related behavior. For example, differences in suckling time, eating speed, food preferences, and crying frequency manifest at birth and correlate strongly with genetics, suggesting that such behaviors are hardwired before the onset of environmental influences [36]. 24 While twin studies, as well as genetic linkage and familial aggregation analyses, demonstrate a strong genetic contribution to obesity, other methodologies are required to identify specific thrifty genes. Genome-wide association analysis (GWAS) involves DNA sequencing of the genome in thousands of humans to identify single nucleotide polymorphisms (SNPs) followed by correlation of SNP prevalence with phenotypic and clinical characteristics. GWAS permits correlation of specific DNA SNPs with clinical and disease-­ related traits. While powerful, a number of issues currently limit the utility of these techniques. First is the aforementioned polygenic nature of obesity. Over 200 specific genetic mutations are associated with obesity in mice and over 50 candidate genes have been identified in humans, likely only a minority of the total genes involved. These polymorphisms are found throughout the genome in and near genes associated with multiple physiologic processes, but the individual effect size of each locus is weak, contributing less than 1–3% to the tendency towards obesity and confounding analysis of the functional role of any single gene on global phenotype. Tens of thousands of subjects are required to adequately power studies to identify SNPs with such low effect magnitudes. In addition, multiple loci interact in concert, further increasing complexity and confounding assignment of specific functions. Finally, many SNPs lie in noncoding regulatory regions of the genome, areas that are currently poorly understood and present challenges with respect to interrogation and functional analysis. Despite these limitations, GWAS has begun to identify thrifty gene candidates. Among these emerging candidate genes, MC4R represents a singularly important locus. Over 90 polymorphisms have been identified within the human MC4R gene locus which together account for up to 6% of cases of common human obesity and are transmitted in monogenic Mendelian fashion with incomplete penetrance [37]. Study of similar high-frequency SNP loci has the potential to identify important markers and mechanisms of disease. Future challenges include identifying and functionally defining other coding and noncoding SNPs and quantifying their contribution to obesity. Metabolic Diversity If we all share the same tightly regulated weight control mechanisms, then why are some lean but others obese? The multiple thrifty polymorphisms in metabolic genes scattered throughout our genomes combine to cause subtle regulatory and functional differences in the many proteins that carry out the tasks of metabolism, providing our species with broad metabolic diversity [38, 39]. Metabolic diversity explains the heterogeneity of the obesity phenotype, with variability in onset and triggers (e.g., adolescence, pregnancy, menopause), severity, anatomic site of excess adiposity, association with metabolic disease, the capacity to lose weight in response to diet, exercise, and surgical therapy and the body R. W. O’Rourke mass index (BMI) range in which individuals exist. The obesity phenotype is heterogeneous because its underlying mechanisms are equally so. This has led some to propose the term obesities [40]. Why did metabolic diversity evolve in humans? First and foremost, we are not a niche species. We inhabit virtually every environment on the globe, and our metabolic heterogeneity allows us to weather differing environments. For example, in their native environment, Inuit Eskimos maintain lower serum LDL levels, higher HDL levels, and lesser degrees of insulin resistance compared with non-Inuit populations despite a high fat diet and higher degree of obesity, suggesting that adaptive genes designed to manage their unique diet were selected for in the Inuit genome [41]. South Pacific Asians are thought to have a high prevalence of insulin resistance due to selection of genes that prevented hypoglycemia during fasting associated with long ocean voyages [42]. Metabolic diversity provides a genetic savings bank that allows for rapid selection of thrifty alleles over a few generations during famine or emigration to new environments, a strength of our species and one of the reasons that we have populated the planet. Environmental and Epigenetic Contributors nvironmental Influences, Obvious and Subtle E Despite the importance of genetics, environment plays a central role in obesity pathogenesis. Dominant among environmental contributors is the shift to modern processed food. Processed food is ubiquitous, plentiful, and cheap; contains high levels of refined sugar, fat, and salt; is calorie-dense and highly palatable; and is engineered to appeal to sensory, physiologic, and emotional inputs. Marketing strategies and food industry, lobbyist, and subsidy policies that maintain accessibility and affordability of the very foods that contribute most to obesity positively reinforce consumption. The dramatic pan-societal decrease in physical activity exacerbates the problem. Our Paleolithic ancestors consumed 30% more calories than modern humans but engaged in much higher levels of physical activity [43]. Human physical activity levels decreased but remained much higher over past centuries than in the last, during which time most humans engaged in regular strenuous physical activity in the context of agriculture: over 40% of the US population engaged in farming before 1900, compared to less than 2% currently [44], and the trend of progressively decreasing physical activity continues. These problems are exacerbated by disruption of circadian rhythms. Physical activity during our evolution cycled in response to circadian rhythms and feast-famine cycles of early hunter-gatherer and agrarian societies in much the same way leptin levels cycle though allostatic deviation. Such cycling is characteristic of multiple physiologic and 2 The Pathophysiology of Obesity and Obesity-Related Disease behavioral processes, disruption of which contributes to obesity. Temporal eating patterns regulate endocrine physiology and can be entrained: providing an extra meal beyond energy requirements to rodents and humans for a period of time will lead to an anticipatory orexigenic ghrelin surge 1 h prior to the scheduled meal and is associated with increased food intake and weight gain [45]. This periodicity extends to the cellular level, as adipocyte proliferation and other metabolic and physiologic processes exhibit similar cycling [46]. Circadian cycling of physical activity, sleep, and eating patterns are disrupted in modern society. The contribution of these disruptions to obesity is evidenced by the correlation between night work, obesity, and metabolic disease. Less obvious factors contribute. Indoor temperatures have increased and stabilized over past decades; sleep patterns are disrupted, and as the obesity epidemic progresses, assortative mating patterns magnify its growth [47]. Environmental influences are complex, powerful, and entrenched. A lucrative food industry and a lack of societal resources to encourage healthy food and physical activity choices remain obstacles to environmental manipulation. Nonetheless, strong correlation between community food and activity resources, obesity, and metabolic disease are observed in multiple studies, and an emerging field of social and environmental engineering has evolved to address this problem [48]. Epigenetics The phrase “nature versus nurture” suggests dichotomous processes, but in fact the very the foundation of Darwinian theory postulates that genetics and environment are intimately linked. Epigenetic regulation of the genome mediates this link, providing a mechanism for rapid genetic responses to environmental stimuli on timescales less than those required for eons of Darwinian evolution by random spontaneous mutation. Epigenetics is defined as post-fertilization covalent modification of the genome independent of DNA base-pair mutations and includes DNA methylation, glycosylation, and myristoylation, along with modification of histones, other DNA-associated proteins, and coding and noncoding RNA that regulate DNA function. Epigenetic modifications occur in response to environmental stimuli and are transmitted for one, two, or more generations. Epigenetic modification of the genome is active during fetal, neonatal, and to a lesser extent adult development in all cells and organisms and has profound effects on gene expression, function, and phenotype. In 1944 during World War II, the Germans cut off food supplies to the Netherlands, and for 1 year Dutch citizens starved. The children of women who gave birth during the “Dutch Hunger Winter” manifested increased prevalence of metabolic disease and obesity as adults, whereas children of the same mothers born before or after the famine manifested a much lower prevalence of metabolic disease [49]. 25 Epigenetics Genetics Environment Physiology Behavior Phenotype Fig. 2.5 Complex interactions dictate phenotype: Epigenetic mechanisms mediate environmental effects on genetics. Genetics has diverse effects on physiology and behavior. Through our behavior we alter our environment, and our environment in turn has diverse effects on our physiology Environmental stimuli “programmed” metabolism in utero independent of classical Darwinian genetics. Excess nutrition during pregnancy has similar effects. The risk of adult metabolic disease and obesity is increased over twofold in people whose mothers had gestational diabetes during their pregnancy compared to siblings born of the same mothers during nondiabetic pregnancies [50]. Murine and primate models demonstrate similar effects of maternal overfeeding on metabolic disease in progeny, demonstrating epigenetic modifications that persist generations. The similar epigenetically mediated metabolic outcome to both maternal overnutrition and undernutrition may represent an adaptive response to the fetus “sensing” instability in external environmental food supplies, whether it be excess or scarcity, leading to rapid epigenetic regulation of the metabolic genome. The thrifty phenotype hypothesis defines epigenetic regulation of metabolism, in contrast to Neel’s thrifty genotype hypothesis, which is based on classical Mendelian genetic transmission of thrifty SNPs. Epigenetic regulation provides a mechanism by which environmental stimuli rapidly alter the genome to prepare offspring for a dynamic environment. Epigenetics mediates the link between environment and genetics, an interaction that determines our physiology and behavior, through which we in turn modify our environment (Fig. 2.5). Pathophysiology of Obesity-Related Disease utrient Excess, Cell Stress, and the Central N Role of Adipose Tissue besity: More than Mass O Obesity is associated with a spectrum of pathology collectively referred to as metabolic disease. Metabolic disease 26 involves all organ systems, reflecting the fact that the systems that regulate energy balance are fundamental to biology. Large epidemiologic studies document dose-dependent increasing risk ratios for metabolic diseases and long-term mortality with increasing BMI. Despite its strength, however, the correlation between obesity and metabolic disease is imperfect. Within the obese population, metabolic disease spans a heterogeneous spectrum. Overweight or lean subjects may suffer metabolic syndrome, while very obese patients may have minimal disease. Ethnicity also influences disease risk independent of weight, testament to the role of genetics in the pathogenesis of metabolic disease. This variability in metabolic disease risk susceptibility is based in complex variable responses of multiple tissues, including adipose tissue, to excess nutrient delivery. Obesity-related diseases were once thought to be the result of mechanical stress on tissues. Indeed, mechanical stress contributes to osteoarthritis, sleep apnea, and venous stasis disease, but even these pathologies result in part from other mechanisms; for example, inflammation contributes to the pathogenesis of sleep apnea and osteoarthritis. Mechanical stress plays little if any role in the pathogenesis of other metabolic diseases such as asthma, atopy, allergy, and cancer, all of which are increased in obesity. Metabolic diseases also develop in lean subjects albeit at lower incidences. So while mechanical stress contributes, it does not provide the whole story. Rather, metabolic disease results from nonmechanical stress that occurs at the cellular level in response to nutrient excess. These stress responses begin in adipose tissue. Adipose Tissue Biology Adipose tissue was once thought of as a homogenous tissue designed primarily for lipid storage but, in the past few decades, has emerged instead as a complex metabolic, endocrine, and immune organ. The adipose tissue-gut-central nervous system interaction regulated by leptin serves as an example of inter-organ communication mediated by adipose tissue. White adipose tissue (WAT) of mesodermal origin comprises the majority of human adipose tissue. WAT resides in discrete anatomic depots each with distinct phenotypes and functions. Obese humans differ in sites of WAT accumulation, with gynecoid distribution defined by excess subcutaneous adipose tissue (SAT) in the hips and buttocks, android distribution characterized by excess intra-abdominal visceral adipose tissue (VAT), and a spectrum of intermediate phenotypes. Excess WAT in general correlates strongly with metabolic disease and long-term mortality, but these risk ratios are magnified at least twofold for excess VAT compared to SAT. Reinforcing its importance, VAT but not SAT lipectomy in obese mice ameliorates diabetes [51]. Omentectomy as an adjunct to bariatric surgery in humans in contrast does not ameliorate metabolic disease, likely due to the greater R. W. O’Rourke degree of complexity of human anatomic adipose tissue depots and the fact that the omentum comprises a lesser percentage of total VAT mass in humans. A unique aspect of VAT is its direct anatomic communication with the liver via the portal venous system, which might explain its detrimental effects without invoking other mechanisms. Important qualitative differences in metabolism in VAT have been described, however, including higher levels of inflammation, lipolysis, β-adrenergic receptor expression, steroid sensitivity, insulin resistance, and adipocyte proliferation and differentiation. These complexities are compounded by the existence of distinct and functionally less well-defined subdepots within VAT and SAT, including omental, retroperitoneal, mesenteric, and perivisceral subdepots for VAT and deep, superficial, truncal, and extremity subdepots for SAT. In addition, adipocytes are not limited to canonical anatomic depots but reside in virtually all tissues including the bone marrow, perivascular and perivisceral tissues, and all subcutaneous locations. Brown adipose tissue (BAT) constitutes a minority of total adipose tissue stores and is associated with increased thermogenic capacity and beneficial effects on systemic metabolism. BAT has been identified in adult humans with positron emission tomography scanning in cervical, supraclavicular, paraspinous, mediastinal, and perirenal depots, with increased metabolic activity and thermogenesis induced by cold and adrenergic stimuli. In contrast to WAT, BAT mass correlates inversely with metabolic disease risk, and BAT stores decrease in humans with increasing age and with increasing obesity [52]. Adipose tissue complexity extends to the cellular level. Adipocyte stem cells of multiple phenotypic and functional potentials, including BAT precursors termed beige or brite (brown-in-white) preadipocytes, reside within WAT and give rise to adipocytes with different phenotypes. This phenotypic plasticity is a target for therapy, as human clinical trials of drugs that induce “browning” or “beiging” of adipose tissue are in progress. In addition, adipose tissue is comprised not only of adipocytes but also a stromal-vascular cell fraction (SVF) that includes endothelial cells, preadipocytes, leukocytes, lymphocytes, and other cell types. Over half of the SVF is comprised of leukocytes and lymphocytes, which are a rich source of cytokines, adipokines, and inflammatory mediators. utrient Excess and Cell Stress Responses N Much like oxygen, the chemical properties that make nutrients efficient vehicles for energy storage and transfer also impart a high degree of bioactivity and the capacity to participate in energy-intensive and potentially damaging reactions within cells. To control these bioenergetic molecules, cells have evolved sophisticated machinery that meters nutrient flux and limits exposure of cells to nutrients and metabolites. 2 The Pathophysiology of Obesity and Obesity-Related Disease These protective mechanisms sequester nutrients in highly regulated organelles, including mitochondria and endoplasmic reticulum. Obesity is associated with chronic increased cellular flux of nutrients and metabolites that overwhelms cellular metering processes and induces cell stress responses that downregulate cell function and eventually induce cell apoptosis and adaptive responses designed to protect tissues from individual cells suffering from nutrient toxicity. These stress responses include three related processes: endoplasmic reticulum stress, oxidative stress, and inflammation. Stress responses are universal to all cells, but in obesity, they have their genesis in adipose tissue. Initially, adipocytes respond to excess nutrient delivery with hypertrophy, storing the majority of nutrients in a unilocular cytoplasmic lipid droplet. Hypertrophy is an adaptive response, but as adipocytes grow to diameters exceeding 100 microns, the diffusion distance of oxygen, cellular hypoxia ensues. Measurement of adipose tissue oxygen levels is fraught with technical challenges and conflicting data exist, but studies in obese mice and humans using platinum-based electrodes, in vivo staining with hypoxia tracers, and expression of hypoxia-inducible genes confirm hypoxia in obese adipose tissue [53, 54]. Hypoxia conspires with chronic nutrient excess to trigger cell stress responses. Foremost among these responses is the endoplasmic reticulum stress response. The endoplasmic reticulum (ER) is a complex cell organelle that coordinates protein and lipid synthesis. Within the rough ER, chaperone proteins ensure accurate protein translation and folding during ribosomal translation of RNA to protein. The ER also manages posttranslational protein modification, sorting, and transport, RNA processing and trafficking, and drug metabolism. In orchestrating its many functions, the ER depends on a constant influx of nutrients to keep pace with protein synthesis. In this capacity, the ER acts as a dominant cellular nutrient sensor, adjusting cell physiology to accommodate fluctuations in nutrient resources. The ER integrates cellular responses to innumerable stimuli, including hypoxia, temperature, toxins, and, of course, nutrients. The ER is a central clearinghouse for cellular function, but it can be overwhelmed. If nutrient flux exceeds ER processing capacity, the ER responds with an adaptive stress response designed to “catch up.” Initially this response involves upregulation of chaperone protein synthesis to maintain accurate protein synthesis. If excess nutrient flux persists, then the ER responds by activating the unfolded protein response (UPR), a complex transcriptional program that downregulates global cellular protein expression and cell metabolism. The mitogen-activated protein kinases (MAPK) JNK and p38 are important downstream effectors of the UPR, inducing insulin resistance by regulating the activity of insulin signaling mediators such as insulin receptor and IRS-1. Finally, if nutrient excess persists and the ER cannot maintain proper protein synthesis, then the UPR progresses 27 to induce cell apoptosis. The UPR thus protects cells from manageable increases in nutrient delivery and triggers cell death when excess nutrient load is insurmountable. Oxidative stress is activated when formation of reactive oxygen species (ROS), including superoxides, oxygen and hydroxyl free radicals, and hydrogen peroxide, byproducts of mitochondrial respiration, exceeds cellular antioxidant capacity. Increased ROS formation occurs with nutrient excess. Initially, the oxidative stress response upregulates cellular antioxidant mechanisms, including antioxidant enzymes (e.g., superoxide dismutase, glutathione peroxidase, catalase), scavenging molecules (e.g., ascorbic acid, vitamin D, urate, divalent metal ions), and thioredoxin and glutaredoxin scavenging systems. If increased ROS generation continues, the oxidative stress response proceeds to increase mitochondrial uncoupling to further reduce ROS production. Further ROS production leads to cross talk between oxidative stress and ER stress responses and, eventually, cell apoptosis. Adipocyte apoptosis resulting from chronic cell stress responses generates sterile inflammation within adipose tissue, as an inflammatory cell infiltrate that includes macrophages, T cells, B cells, NK cells, and eosinophils is recruited to scavenge dead and dying adipocytes. Adipose tissue macrophages (ATM) are initial responders and dominant effectors of this inflammatory response, congregating around dead or dying adipocytes in crown-like structures (Fig. 2.6, [55]). ATM comprise 10–15% of adipose tissue leukocytes, are increased in obese adipose tissue, and correlate directly with body weight, with reductions in ATM number observed with weight loss, including after bariatric surgery [56]. ATM express multiple cytokines, including TNF-α, IL-10, IL-6, Fig. 2.6 ATM crown-like structure: Adipose tissue macrophages (brown) surround an apoptotic adipocyte. (From: O’Rourke et al. [55]. Reprinted with permission from Springer Nature) 28 IL-8, and IL-1β, as well as exerting other effector functions, all of which in turn regulate diverse aspects of adipocyte metabolism, inducing insulin resistance and lipogenesis and decreasing lipolysis. The central role of ATM in systemic metabolic disease is demonstrated by experiments in which transgenic adipose tissue-specific knockdown of the macrophage homing molecule CCL2 abrogates insulin resistance in obese mice, while overexpression of CCL2 induces insulin resistance in lean mice [57, 58]. ATM are highly heterogeneous and are shifted towards a pro-inflammatory phenotype in obesity. Obesity-specific pathogenic ATM subpopulations have been identified. The integrin CD11c represents an important marker for diabetogenic ATM. CD11c+ ATM are increased in obese mice and humans and induce insulin resistance in adipocytes in vitro, and in vivo ablation of CD11c+ macrophages in obese mice ameliorates diabetes [59, 60]. Targeting ATM represents an important therapeutic strategy for metabolic disease. ATM are not the only cellular effectors of adipose tissue inflammation. A pan-leukocyte infiltrate is present in obese adipose tissue that includes T cells, B cells, NK cells, and eosinophils, which potentiate ATM inflammatory responses and regulate adipocyte metabolism. Cytotoxic T cells are increased in number in adipose tissue in obese mice and humans, while regulatory T cells, which attenuate macrophage inflammatory responses, are reduced. B cells, NK cells, and eosinophils also regulate adipose tissue inflammation. In addition, adipocytes themselves express inflammatory cytokines and prime ATM inflammatory responses. Finally, as inflammation persists, adipose tissue fibrosis ensues, limiting adipocyte hypertrophic capacity and exacerbating adipocyte failure. Model for Adipose Tissue Dysfunction A Obese adipose tissue manifests multiple signs of cell stress. ER stress mediators are increased in adipose and other tissues from obese humans and reduced with weight loss, including after bariatric surgery. Induction of ER stress in mice induces systemic insulin resistance and steatosis, while inhibition of ER stress has the opposite effect [61]. Increased oxidative stress, ROS levels, and reduced mitochondrial number, size, and function in serum, cells, and tissues from obese humans have been demonstrated using HPLC, mass spectrometry, and other assays [62]. Obese humans mount increased peripheral blood monocyte ROS responses to a meal, suggesting metabolic differences that predispose to oxidative stress [63]. Chemical inducers of mitochondrial uncoupling or ROS scavengers reverse insulin resistance in obese mice, demonstrating the potential for therapy targeting oxidative stress [64]. ER stress, oxidative stress, and inflammation are fundamental cellular processes that potentiate one another with R. W. O’Rourke Nutrient excess, moderate adipocyte hypertrophy Early compensatory ER stress Continued nutrient excess, advanced adipocyte hypertrophy Hypoxia Progressive ER stress, oxidative stress Inflammation Fibrosis, limited adipocyte hypertrophic capacity Late ER stress, UPR, advanced oxidative stress, adipocyte necrosis, apoptosis Failure of adipocyte nutrient buffering capacity, systemic overflow Fig. 2.7 A model for adipose tissue failure in obesity: In early obesity, adipocytes respond to nutrient excess with hypertrophy. As nutrient excess persists, adipocyte hypertrophy continues, leading to hypoxia and ER and oxidative stress responses, causing adipocyte apoptosis and an inflammatory response. Hypoxia, ER and oxidative stresses, and inflammation potentiate one another, inducing a vicious cycle. These processes eventually lead to adipose tissue fibrosis, which further limits adipocyte storage capacity exacerbating failure of adipose tissue nutrient buffering capacity. As adipose tissue failure proceeds, nonesterified fatty acids, other nutrients, metabolites, and inflammatory cytokines and adipokines overflow into the systemic circulation intimate overlap in signaling pathways. While the exact sequence, kinetics, and causal relationships of adipocyte hypertrophy, nutrient excess, hypoxia, and cell stress responses are not yet well defined, these events provide a model for adipose tissue failure (Fig. 2.7). The end result of these processes is impairment of adipose tissue’s nutrient buffering capacity. As adipose tissue nutrient storage capacity is overwhelmed, nutrients “overflow” to peripheral tissues, and metabolic disease metastasizes. 2 The Pathophysiology of Obesity and Obesity-Related Disease eyond Adipose Tissue: Systemic Metabolic B Disease Adipose Tissue Overflow In early obesity, nutrient excess, cell stress, and metabolic dysfunction are confined to adipose tissue. Adipocytes are exquisitely designed to store lipid and tolerate nutrient toxicity, but as nutrient excess persists, their storage capacity is overwhelmed and nutrients overflow to peripheral tissues. Nonesterified free fatty acids are released into the systemic circulation and accumulate in peripheral tissues, and lipotoxicity contributes to peripheral organ dysfunction. Excess glucose and other simple carbohydrates induce formation of advanced glycation end products in multiple tissues. ER stress, oxidative stress, and inflammation unfold in peripheral tissues with mechanisms similar to those described in adipose tissue (Fig. 2.8). VAT drains its venous effluent to the liver via the portal venous system, leading to portal overflow of excess nutrients from VAT to the liver and establishing the liver as a dominant secondary site of aberrant metabolism. Increased free fatty acid delivery leads to hepatic steatosis, inflammation, and insulin resistance. Aspects of the portal hypothesis are debated. For example, isotope dilution techniques demonstrate that only 20% of free fatty acids delivered to the portal circulation derive from VAT, with the majority from recirculation into the splanchnic bed from the systemic circulation, calling into question the magnitude of the effect of VAT on liver metabolism [65]. Nonetheless, the portal hypothesis provides a coherent rationale for VAT’s role in the pathogenesis of liver disease in obesity. As obesity progresses, the Excess caloric intake CNS: Aberrant nutrient, hormonal, neural afferent inputs and efferent outputs Adipose tissue: Hypertrophy, ER, Ox stress, inflammation, overflow Liver: NAFLD, inflammation, insulin resistance Gut: Microbiome derangements Peripheral tissues: Lipotoxicity, inflammation, ER, Ox stress, peripheral insulin resistance Fig. 2.8 Inter-organ communication and systemic metabolic disease: A simplified model for systemic spread of metabolic disease. Excess nutrient intake first affects adipose tissue, but evidence supports direct effects on the CNS and the gut microbiome as well. As adipose tissue dysfunction progresses, overflow to the liver and other peripheral organs induces systemic metabolic disease. The progression of systemic metabolic disease in the liver and other peripheral tissues leads to aberrant hormonal and neural communications between CNS and peripheral organ systems as well as between peripheral organs. Not shown are arrows representing direct communication between the CNS, the gut, and all peripheral organ systems 29 byproducts of adipose tissue and liver metabolism and inflammation overflow into the systemic circulation via peripheral overflow, and all tissues suffer similar insults. Lipotoxicity, ER and oxidative stress, and inflammation are observed in the skeletal muscle, liver, vasculature, lung, and other tissues. The overflow concept underscores the importance of adipose tissue as a buffer for excess nutrients. Adipose tissue’s ability to store large amounts of lipid and other nutrients protects other tissues not designed to tolerate lipotoxicity and nutrient excess. This concept is reinforced by patients with various forms of lipodystrophy who lack adipose tissue and develop severe insulin resistance and hyperlipidemia, similar to obese patients. Adipocyte hypertrophy is an adaptive response which confines the stress induced by nutrient excess in early obesity to adipose tissue. As obesity progresses, however, adipose tissue storage capacity is overwhelmed, and nutrient excess affects other tissues. Systemic metabolic disease results from a complex communication between dysfunctional adipose tissue and all other organ systems. entral Nervous System: Peripheral Organ C Communication The complexities of central nervous system (CNS) regulation of energy homeostasis are beyond the scope of this chapter, but in brief, the brain receives at least three primary types of afferent signals regarding energy homeostasis: dietary constituents, hormones, and neural afferents. Neurons sensitive to dietary constituents, including glucose, free fatty acids, amino acids, and their metabolites, reside primarily in the ARCN and VMH of hypothalamus. Long-­ chain fatty acids (LCFA) are important stimuli: normal efferent signals from LCFA-sensitive neurons act to limit insulin resistance and food intake. Chronic stimulation by saturated LCFA leads to accommodation and exhaustion of LCFA-­ responding hypothalamic neurons, whereas polyunsaturated fatty acids (PUFA) do not cause accommodation, thus explaining the detrimental and beneficial health effects of saturated LCFA and PUFA, respectively. Glucose- and amino acid-sensitive neurons also exist in the hypothalamus and other areas of the brain, and other metabolites also act as afferent signals to the CNS. The brain also receives hormonal afferent signals. Leptin and insulin are dominant, but multiple other hormonal mediators impact on hypothalamic and other CNS networks, including glucagon, CCK, amylin, ghrelin, and others. The hypothalamus also receives sympathetic and parasympathetic neural afferent inputs from multiple organs, including the liver, adipose tissue, gut, viscera, and skeletal muscle. Gut afferents communicate short-term food resource availability, while adipose tissue afferents signal the status of long-term energy resources. Hepatic afferents communicate the status of glucose and fatty acid level resources, and all organs and tissues deliver afferent signals 30 to the brain that provide similar information regarding energy stores. Efferent signals from the brain to the periphery are complex, are poorly defined, and regulate diverse aspects of food-related behavior and metabolism. The hypothalamus generates anorexigenic and orexigenic programs in response to multiple stimuli that involve not only satiety and hunger but behavioral, emotional, physiologic, and reward responses and are mediated by neural efferents to other areas of the brain as well as to all peripheral organ systems. Virtually all afferent CNS inputs are altered in obesity. Hyperglycemia and hyperlipidemia contribute to the many aberrations in metabolite input to the CNS, while hyperinsulinemia and hyperleptinemia, along with multiple other adipokine and gut hormones, alter hormonal inputs. Hypothalamic resistance to leptin and insulin result, further disrupting CNS regulation of energy balance. Peripheral organ dysfunction contributes to aberrant neural afferent inputs to the CNS. Research into central-peripheral neural interactions is in early stages, but such communication is clearly important: overexpression of the hepatic receptor for the adipogenic mediator PPAR-γ2 in mice leads to steatosis and obesity which is abrogated by transection of the hepatic branch of the vagus nerve, demonstrating that afferent signals from liver to brain regulate food intake and metabolism. Inflammation contributes to CNS dysregulation. Obese mice manifest increased hypothalamic levels of the inflammatory cytokines with a concomitant increase in inflammatory signaling. Nutrient excess contributes, as fatty acids generate hypothalamic inflammation and ER stress [66]. Hypothalamic inflammation mediates resistance to leptin’s and insulin’s anorexigenic effects, well-described phenomena in obesity, and blockade of hypothalamic ER stress or inflammatory signaling ameliorates hypothalamic leptin and insulin resistance and attenuates metabolic disease [61, 66]. Many questions remain: for example, direct blockade of inflammatory cytokines within the hypothalamus paradoxically exacerbates rather than attenuates obesity and metabolic disease, suggesting some beneficial effects of hypothalamic inflammation [66]. Nonetheless, this emerging field of study demonstrates promise for manipulation of central mechanisms of metabolism. he Gut Microbiome T The role of the gut microbiome in the pathogenesis of metabolic disease is an important emerging field. “Metagenomic” sequencing of microbiome DNA, analysis of bacterial ribosomal RNA, and microbiota speciation are tools currently used to interrogate the microbiome. Despite significant variation between individuals, gut microbiota speciation within individuals remains constant over time in the absence of changes in energy homeostasis or physiology. Anywhere from 1000 to 30,000 species of bacteria reside within the R. W. O’Rourke human gut, with total body bacterial number an order of magnitude greater than total body cell number. Commensal gut microbiota play an important role in metabolism, potentiating digestion and absorption of dietary constituents. Animals raised in germ-free environments have 40–60% lower body weight despite higher food intake relative to those raised in standard conditions, and stool transfer from mice with a normal complement of gut microbiota increases body fat in recipient germ-free animals. Furthermore, stool transfer from obese animals leads to greater weight gain compared to stool from lean animals, suggesting that alterations in the microbiome contribute to obesity [67]. Indeed, gut microbiota speciation is altered in obesity. Over 90% of gut microbiota fall into the phyla Bacteroidetes or Firmicutes; obesity in rodents and humans is associated with an increased Firmicutes/Bacteroidetes ratio that is reversed with weight loss, including after bariatric surgery, and probiotics accelerate diet-induced and surgically induced weight loss. Gut microbiota regulate systemic metabolism and contribute to metabolic disease. Microbiota speciation patterns correlate with metabolic disease in humans and mice. Fecal transplant experiments in mice and humans demonstrate that gut microbiota are capable of transferring obese, lean, and metabolically diseased and healthy phenotypes. Underlying mechanism is not well defined, but inflammation is implicated: commensal gut bacteria regulate immune function and prevent colonization by pathogenic bacteria. Derangements in microbiota induce inflammation through activation of pattern recognition receptors such as Toll-like receptors. Obesity in mice and humans is associated with increased gram-­ negative gut bacteria and increased absorption of lipopolysaccharide [68], and antibiotic therapy directed towards gram-negative bacteria decreases gut luminal lipopolysaccharide concentrations, systemic inflammation, and hepatic steatosis in rats [69]. Manipulation of gut microbiota represents an important therapeutic frontier for metabolic disease. Specific Metabolic Diseases Liver Disease Nonalcoholic fatty liver disease (NAFLD) defines a range of pathology that includes hepatic steatosis, steatohepatitis, and cirrhosis. NAFLD is primarily a disease of obesity but may exist in lean patients. Hepatic steatosis, defined as hepatic lipid content >95th percentile for healthy lean subjects or alternatively, as >5% of hepatocytes with cytoplasmic lipid, is present in 15–30% of the general population depending on diagnostic modality and in over 90% of patients with BMI >40 [70]. Up to 30% of subjects with steatosis will progress to steatohepatitis, a histologic diagnosis defined by hepato- 2 The Pathophysiology of Obesity and Obesity-Related Disease cyte ballooning, apoptosis, inflammation, Mallory bodies, and/or fibrosis. Of patients who develop steatohepatitis, 10–30% will progress to cirrhosis within a decade. Accurate noninvasive predictors of disease progression remain elusive and represent an active area of research. Liver biopsy is the current gold standard for diagnosis. Steatosis and steatohepatitis are influenced by genetics. Ethnic subpopulations demonstrate variable propensity to NAFLD, with Asians and Hispanics at highest risk, Caucasians at intermediate risk, and African Americans at lowest risk. Histology also varies among ethnicities: Hispanics demonstrate increased Mallory bodies, while hepatocyte ballooning predominates in Asians, suggesting different mechanisms of pathogenesis [71]. NAFLD is highly hereditable and NAFLD-associated polymorphisms are identified in humans [72]. Steatosis results from an imbalance in hepatic lipid uptake and release. Three primary sources of hepatic lipid include (1) dietary lipid from the intestine as packaged as triglycerides in chylomicrons that are subsequently hydrolyzed into nonesterified free fatty acids and delivered to the liver bound to albumin; (2) de novo hepatic lipogenesis from glucose and fructose substrates, regulated by PPAR-γ, C/EBPα, and SREPB1, transcription factors that regulate cellular lipogenesis in adipocytes and hepatocytes; and (3) free fatty acid delivery from adipose tissue. In obese subjects, isotope labeling studies demonstrate that approximately 60% of hepatic lipid is derived from free fatty acids from adipose tissue, 25% from de novo lipogenesis, and 15% from diet [73]. Hepatic lipid uptake is balanced by lipid export via VLDL synthesis. Obesity is associated with derangements in all aspects of hepatic lipid flux. Increased dietary carbohydrates, especially fructose, increase hepatic de novo lipogenesis via activation of SREPB1. In addition, VLDL export is compromised in obesity in part secondary to decreased expression of hepatic apolipoprotein B [74]. Nonetheless, increased free fatty acid delivery from excess adipose tissue is the dominant source of hepatic lipid in obesity. Steatosis correlates closely with hepatic and systemic insulin resistance, both of which are exacerbated due to the selective nature of insulin resistance in hepatocytes, which become resistant to insulin’s normally suppressive effects on gluconeogenesis, while at the same time remaining sensitive to insulin-mediated stimulation of de novo lipogenesis. Selective hepatic insulin resistance thus promotes both hyperglycemia and steatosis. As steatosis progresses, excess free fatty acids and their derivatives, including ceramide and diacylglycerols, induce ER and oxidative stress, and eventually apoptosis in hepatocytes, generating an inflammatory response that contributes to NAFLD. Expression of pro-inflammatory cytokines, including TNF-α and IL-6, is increased, inducing steatohepatitis. Neutralization of TNF-α in obese mice reduces steatohepatitis, and anti-TNF-α antibody ameliorates steatosis in humans, 31 suggesting the possibility of cytokine-based therapy for NAFLD [75]. Similar to adipose tissue, hepatic macrophage (Kupffer cell) infiltration is increased, while anti-­ inflammatory NKT cells are reduced. The MAPK JNK is a nexus for cell stress in hepatocytes, is activated by inflammation, free fatty acids, and ER and oxidative stress, and in turn generates hepatic insulin resistance. Hepatic JNK activity is increased in obese mice, while JNK knockout mice are protected from steatohepatitis and insulin resistance [76]. Finally, alterations in the microbiome, intestinal epithelial integrity, and increased hepatic endotoxin exposure have been implicated in NAFLD [77]. Current treatment for NAFLD is limited to diabetes control, weight loss, and exercise. Bariatric surgery ameliorates steatosis and steatohepatitis. Thiazolidinediones and antioxidant therapy are emerging treatment modalities, but long-term efficacy and safety are not yet defined. Diabetes By 2050, it is estimated that a third of the US population will be afflicted by type II diabetes [78]. Obesity is a strong risk factor for diabetes, with odds ratios exceeding 5 [79]. Peripheral insulin resistance underlies the pathogenesis of diabetes. Insulin binds its receptor on target cells and initiates an intracellular signaling cascade involving activation of insulin receptor substrate-1 (IRS-1) and Akt, which in turn mediate insulin’s effects on glucose homeostasis and in addition transmit parallel signals that induce cell proliferation and inflammation. Skeletal muscle is responsible for 70–80% of glucose utilization and is a dominant site of peripheral insulin resistance. Increased lipid delivery to skeletal muscle leads to downregulation of molecules involved in myocyte glucose uptake as muscle shifts from glucose to lipid metabolism. This decrease in muscle glucose utilization contributes to hyperglycemia, which in turn induces a compensatory increase in pancreatic beta-cell insulin secretion. Hyperinsulinemia induces increased proliferation, increased collagen synthesis, and activation of JNK in myocytes, all of which potentiate skeletal muscle insulin resistance creating a vicious cycle. Inflammation also contributes, with increased skeletal muscle macrophage infiltration and inflammatory cytokine expression [80]. The inflammatory cytokines TNF-α and IL-6 promote skeletal muscle insulin resistance, although IL-6 may have the opposite effect during exercise [81]. The anti-inflammatory mediators IL-10 and adiponectin are decreased in obesity and attenuate skeletal muscle insulin resistance [80]. In early insulin resistance, β-cell mass and insulin secretion increase leading to compensatory hyperinsulinemia which delays the onset of hyperglycemia. As obesity and insulin resistance progress, ER and oxidative stress and inflammation are triggered by free fatty acids and glucotoxicity and cause beta-cell failure and eventually death. 32 Salsalate, a modern anti-inflammatory salicylate derivative, is being studied in humans as treatment for diabetes [82]. Initial trials of TNF-α antibody therapy in diabetic humans did not demonstrate therapeutic efficacy, but recent data from diabetic patients treated with long-term anti-­TNF-­α therapy for autoimmune diseases demonstrate improved glucose homeostasis and have reinvigorated interest in TNF-α blockade [83]. Anti-IL-1 antibody therapy also shows promise as treatment for diabetes in early human clinical trials [84]. The importance of macrophages in insulin resistance has prompted clinical trials in diabetic humans of drugs designed to block macrophage homing to adipose tissue. Vascular Disease Risk ratios for atherosclerotic disease, hypertension, and dyslipidemia associated with obesity range from 1.5 to 3.0 [79]. Vascular disease is closely linked to obesity and type II diabetes and a dominant cause of mortality in diabetic patients. The importance of insulin in the pathogenesis of atherosclerosis is demonstrated by experiments in animals in which arterial infusion of insulin induces atherosclerosis in the infused arteries [85]. Hyperinsulinemia stimulates proliferation and collagen synthesis in arterial smooth muscle cells via Akt-mediated signaling pathways that are preserved in insulin resistance as discussed above. Insulin also increases LDL transport into arterial smooth muscle cells, an early event in atherosclerosis that causes lipotoxicity with resultant ER stress, oxidative stress, and inflammation. Macrophages and T cells infiltrate atheromas and contribute to plaque formation and progression. Saturated fatty acids and derivatives directly activate TLR4 on these cells, exacerbating the inflammatory response. Nitric oxide (NO) plays a central role in vascular disease in obesity. NO has vasorelaxant, anti-inflammatory, antioxidant, and antiproliferative action. Insulin’s normal Akt-mediated stimulatory effect on endothelial NO synthase is downregulated in insulin resistance, reducing NO levels. Free fatty acids and ROS from oxidative stress also directly inactivate NO. Insulin-resistant mice demonstrate decreased NO and increased atherosclerosis. NO production is decreased and abnormalities in NO-mediated vasodilation correlate with coronary artery disease in diabetic humans [86]. Multiple pharmacologic agents are used to treat vascular disease, many of which mediate their effects in part by attenuating inflammation, most notably statins. Novel therapy for vascular disease directed towards inhibiting oxidative stress, leukotriene synthesis, phospholipases, and inflammatory cytokines is in human clinical trials [87]. Cancer Obesity is associated with an increased risk of cancer. Inflammation promotes carcinogenesis, and the association between cancer and inflammation independent of obesity is well-established. Inflammation is generally associated with catabolic states, such as sepsis, cachexia, and chronic illness. R. W. O’Rourke Obesity, in contrast, is one of the few inflammatory states associated with anabolic signaling, which in the context of chronic inflammation and increased cell stress and turnover increases the opportunity for mutagenesis but in the absence of the catabolic brake on proliferation that normally prevents propagation of such damage. This combination of inflammation and anabolism is a perfect storm for carcinogenesis. Diabetes is independently associated with increased cancer risk, including breast, pancreatic, hepatic, endometrial, colorectal, and kidney. Insulin is a growth factor for multiple cell types and binds not only the insulin receptor but also the IGF-1 receptor to generate a pro-proliferative program. Insulin receptor expression is upregulated in many cancers, supporting a tropic role for insulin in tumor growth. Insulin also induces hepatic growth hormone receptor expression, which in turn increases growth hormone-mediated hepatic IGF-1 expression. IGF-1 also acts as a growth factor for many cells, including tumor cells, and increased IGF-1 levels are associated with increased risk of cancer. Other adipokines and cytokines also regulate tumor proliferation. Leptin has growth-promoting effects on cancer cells in vitro, and hyperleptinemia is a risk factor for certain cancers [88]. Adiponectin, levels of which are decreased in obesity, has a protective effect, inhibiting tumor growth in vitro, while increased adiponectin levels have been linked to lower cancer risk in epidemiologic studies. Altered steroid metabolism provides further proliferative stimulus. Estrogen receptor expression is increased in multiple cancers, and obesity is associated with increased estrogen levels as a result of peripheral conversion of androgens to estrogens via aromatase expressed by adipose tissue. Furthermore, insulin and IGF-1, both increased in obesity, inhibit sex hormone-­binding globulin expression, increasing steroid bioavailability [89]. Hyperglycemia and lipotoxicity contribute to carcinogenesis. Tumor cells avidly take up glucose and are adapted to anaerobic energy production (the Warburg effect), providing a growth advantage but increasing oxidative stress. Saturated free fatty acids promote tumor growth, as do adipocytes themselves. Peri-tumor adipocytes are associated with worse outcomes in multiple cancers, including the prostate, kidney, colon, and breast. Peri-tumor adipocytes produce cytokines and adipokines that upregulate proliferative genes and downregulate apoptotic programs in tumors, as well as matrix metalloproteases implicated in metastasis. Adipocytes have been shown to deliver metabolites, including fatty acids and glutamine, to tumor cells, engaging in metabolic cross talk that essentially “feeds” tumor cells and promotes tumor cell proliferation. The intracellular signaling kinase Akt provides an example of the overlap between metabolism and carcinogenesis. Akt regulates multiple aspects of cell physiology and is one of the most commonly mutated genes in cancer. Akt regulates glucose homeostasis and cell proliferation through distinct signaling pathways. Insulin activates Akt, and insulin 2 The Pathophysiology of Obesity and Obesity-Related Disease resistance in obesity is selective with respect to Akt and characterized by resistance to insulin’s effects on glucose homeostasis but preservation of insulin-triggered Akt-mediated activation of mitogenic and inflammatory signaling [90]. The hypoglycemic agents metformin and thiazolidinediones inhibit Akt’s proliferative effects while potentiating insulin signaling activity, and long-term treatment with these drugs in diabetic humans may be associated with decreased cancer risk [91], suggesting the potential for cancer therapy by exploiting metabolic drugs. Conclusion 33 Question Section Part I 1. The genetic contribution to the obesity phenotype based on twin studies is estimated to be: A. <1% B. 10% C. 50% D. 70% 2. Thermogenesis: A. Occurs when mitochondrial oxidative phosphorylation is completely coupled to electron transport B. Is likely to be increased in obese subjects C. Is increased with increased uncoupling protein activity D. Is decreased in response to a meal 3. Leptin: A. Secretion is decreased in response to a meal B. Is secreted by the hypothalamus and binds its receptors in the gut to induce satiety C. Induces satiety and has primarily pro-inflammatory effects D. Induces endoplasmic reticulum stress The above review of specific metabolic diseases is by necessity incomplete. Obesity is associated with kidney disease, asthma, atopy, allergic disease, endocrine and cognitive disorders, and a range of other pathologies. The processes that unfold in the tissues in which these diseases arise parallel those discussed above in adipose tissue, liver, and skeletal muscle. An understanding of this pathophysiology will lead to therapy for metabolic disease. Conceptually, therapy may be directed towards obesity itself or its metabolic sequelae. Manipulation of satiety and hunger factors to prevent or reverse obesity is a dominant effort and includes MC4R agonists and antagonists, cannabinoid antagonists, and anti-­ ghrelin “vaccines.” Drugs in various stages of research Part II designed to increase energy expenditure regulate mediators of cellular energy homeostasis, such as PPAR-δ, SIRT1, 4. Endoplasmic reticulum stress: APMK, and dopamine receptors, among others. Methods to A. Results from excess nutrient delivery to cells as well manipulate adipocyte metabolism also hold promise. as multiple other stimuli Multiple agents that induce “browning” of WAT to a metaB. Is primarily mediated by reactive oxygen species bolically favorable BAT phenotype are in human clinical triC. Occurs only in adipocytes als. Manipulation of oxidative, stress, ER stress, and D. Occurs in the hypothalamus in response to leptin MAPK-related mediators are among the many avenues of bonding its receptors research towards this goal. Recent data demonstrating that 5. Insulin resistance: hypothalamic leptin resistance may be ameliorated by agents A. Is selective in the liver, leading to increased gluconeothat reduce ER stress has reinvigorated interest in leptin thergenesis and increased de novo lipogenesis apy [61]. Inflammation-based therapies including salsalate, B. Is selective in the liver, leading to decreased glucocytokine-directed therapies, and macrophage homing-­ neogenesis and decreased de novo lipogenesis directed strategies are under active study in human trials. C. Is characterized in early diabetes by pancreatic beta-­ Manipulation of macrophage phenotype and function and cell failure and a defect in insulin secretion targeting of specific pathogenic macrophage subpopulations D. Is characterized by increased IRS-1 activity represents an important avenue of research. Targeting other 6. Adipose tissue overflow: cells involved in adipose tissue inflammation, such as regulaA. Is a normal response to a meal tory T cells, NK cells, and NKT cells, has also shown potenB. Results from a failure of adipose tissue nutrient bufftial in animal models. Obesity-related metabolic disease ering capacity and underlies systemic metabolic exacts an enormous toll on public health, and an understanddisease ing of its pathophysiology holds promise for novel pharmaC. Is characterized by reduced free fatty acid delivery to cotherapy or genetic therapies for a wide range of diseases the liver with enormous impact on the human condition. D. Leads to decreased systemic cellular stress 34 References 1. Friedman JM. Modern science versus the stigma of obesity. Nat Med. 2004;10(6):563–9. 2. Prentice AM. Fires of life: the struggles of an ancient metabolism in a modern world. Br Nutr Found Nutr Bull. 2001;26:13–27. 3. Conard NJ. A female figurine from the basal Aurignacian of Hohle Fels Cave in southwestern Germany. Nature. 2009;459:248–52. 4. Flegal KM, Carroll MD, Ogden CL, Curtin LR. Prevalence and trends in obesity among US adults. JAMA. 2010;303(3):235–41. 5. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372(6505):425–32. 6. Farooqi IS, Jebb SA, Langmack G, Lawrence E, Cheetham CH, Prentice AM, et al. Effects of recombinant leptin therapy in a child with congenital leptin deficiency. N Engl J Med. 1999;341:879–84. 7. Considine RV, Sinha MK, Heiman ML, Kriauciunas A, Stephens TW, Nyce MR, et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N Engl J Med. 1996;334:292–5. 8. Heymsfield SB, Greenberg AS, Fujioka K, Dixon RM, Kushner R, Hunt T, et al. Recombinant leptin for weight loss in obese and lean adults: a randomized, controlled, dose-escalation trial. JAMA. 1999;282:1568–75. 9. Crowley VE. Overview of human obesity and central mechanisms regulating energy homeostasis. Ann Clin Biochem. 2008;45(Pt 3):245–55. 10. Berridge KC, Ho CY, Richard JM, DiFeliceantonio AG. The tempted brain eats: pleasure and desire circuits in obesity and eating disorders. Brain Res. 2010;1350:43–64. 11. Zarkesh-Esfahani H, Pockley G, Metcalfe RA, Bidlingmaier M, Wu Z, Ajami A, et al. High-dose leptin activates human leukocytes via receptor expression on monocytes. J Immunol. 2001;167(8):4593–9. 12. Paz-Filho G, Babikian T, Asarnow R, Esposito K, Erol HK, Wong M, et al. Leptin replacement improves cognitive development. PLoS One. 2008;3(8):1–7. 13. Kim W, Egan JM. The role of incretins in glucose homeostasis and diabetes treatment. Pharmacol Rev. 2008;60(4):470–512. 14. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science. 1993;259(5091):87–91. 15. Briggs DI, Andrews ZB. A recent update on the role of ghrelin in glucose homeostasis. Curr Diabetes Rev. 2011;7(3):201–7. 16. Leibel RL, Rosenbaum M, Hirsch J. Changes in energy expenditure resulting from altered body weight. N Engl J Med. 1995;332:621–8. 17. Rosenbaum M, Vandenborne K, Goldsmith R, Simoneau J, Heymsfield S, Joanisse D, et al. Effects of experimental weight perturbation on skeletal muscle work efficiency in human subjects. Am J Phys. 2003;285:R183–92. 18. Astrup A, Gotzsche PC, van Werken K, Ranneires C, Toubro S, Raben A, et al. Meta-analysis of resting metabolic rate in formerly obese subjects. Am J Clin Nutr. 1999;69:1117–22. 19. Stock MJ. Gluttony and thermogenesis revisited. Int J Obes. 1999;23:1105–17. 20. Bouchard C, Tremblay A, Despres JP, Nadeau A, Lupien PJ, Theriault G, et al. The response to long-term overfeeding in identical twins. N Engl J Med. 1990;322:1477–82. 21. Straznicky NE, Eikelis N, Nestel PJ, Dixon JB, Dawood T, Grima MT, et al. Baseline sympathetic nervous system activity predicts dietary weight loss in obese metabolic syndrome subjects. J Clin Endocrinol Metab. 2012;97(2):605–13. 22. Rosenbaum M, Hirsch J, Murphy E, Leibel RL. Effects of changes in body weight on carbohydrate metabolism, catecholamine excretion, and thyroid function. Am J Clin Nutr. 2000;71(6):1421–32. R. W. O’Rourke 23. Levine JA, Vander Weg MW, Hill JO, Klesges RC. Non-exercise activity thermogenesis: the crouching tiger hidden dragon of societal weight gain. Arterioscler Thromb Vasc Biol. 2006;26(4):729–36. 24. Enerback S. Brown adipose tissue in humans. Int J Obes. 2010;34:S43–6. 25. Rothwell NJ, Stock MJ. A role for brown adipose tissue in diet-­ induced thermogenesis. Nature. 1979;281:31–5. 26. Dulloo AG, Samec S. Uncoupling proteins: do they have a role in body weight regulation? News Physiol Sci. 2000;15:313–8. 27. Kopecky J, Clarke G, Enerback S, Spiegelman B, Kozak LP. Expression of the mitochondrial uncoupling protein gene from the aP2 gene promoter prevents genetic obesity. J Clin Invest. 1995;96:2914–23. 28. Musa CV, Mancini A, Labruna G, Valerio G, Franzese A, Pasanisi F, et al. Four novel UCP3 gene variants associated with childhood obesity: effect on fatty acid oxidation and on prevention of triglyceride storage. Int J Obes. 2012;36(2):207–17. 29. Gesta S, Tseng YH, Kahn CR. Developmental origin of fat: tracking obesity to its source. Cell. 2007;131(2):242–56. 30. Bessesen DH. Regulation of body weight: what is the regulated parameter? Physiol Behav. 2011;104:599–607. 31. Ostan I, Poljsak B, Simcic M, Tijskens LMM. Appetite for the selfish gene. Appetite. 2010;54:442–9. 32. Neel JV. Diabetes mellitus: a “thrifty” genotype rendered detrimental by “progress”? Am J Hum Genet. 1962;14(4):353–62. 33. Speakman JR. Thrifty genes for obesity, an attractive but flawed idea, and an alternative perspective: the ‘drifty gene’ hypothesis. Int J Obes. 2008;32(11):1611–7. 34. Speakman JR. A nonadaptive scenario explaining the genetic predisposition to obesity: the “predation release” hypothesis. Cell Metab. 2007;6(1):5–12. 35. McEvoy BP, Powell JE, Goddard ME, Visscher PM. Human population dispersal “Out of Africa” estimated from linkage disequilibrium and allele frequencies of SNPs. Genome Res. 2011;21(6):821–9. 36. Llewellyn CH, van Jaarsveld CH, Johnson L, Carnell S, Wardle J. Nature and nurture in infant appetite: analysis of the Gemini twin birth cohort. Am J Clin Nutr. 2010;91(5):1172–9. 37. Loos RJ, Lindgren CM, Li S, Wheeler E, Zhao JH, Prokopenko I, et al. Common variants near MC4R are associated with fat mass, weight and risk of obesity. Nat Genet. 2008;40(6):768–75. 38. Hawkes J, Wang ET, Cochran GM, Harpending HC, Moyzis RK. Recent acceleration of human adaptive evolution. PNAS USA. 2007;104(52):20753–8. 39. Teebi AS, Teebi SA. Genetic diversity among the Arabs. Community Genet. 2005;8(1):21–6. 40. Guy-Grand B. From obesity to obesities: from concepts to practices. Ann Endocrinol (Paris). 2003;64(5 Pt 2):S7–15. 41. Voruganti VS, Cai G, Cole SA, Freeland-Graves JH, Laston S, Wenger CR, et al. Common set of genes regulates low-density lipoprotein size and obesity-related factors in Alaskan Eskimos: results from the GOCADAN study. Am J Hum Biol. 2006;18(4):525–31. 42. Myles S, Lea RA, Ohashi J, Chambers GK, Weiss JG, Hardouin E, et al. Testing the thrifty gene hypothesis: the Gly482Ser variant in PPARGC1A is associated with BMI in Tongans. BMC Med Genet. 2011;12:10. 43. Hayes M, Chustek M, Heshka S, Wang Z, Pietrobelli A, Heymsfield SB. Low physical activity levels of modern Homo sapiens among free-ranging mammals. Int J Obes. 2005;29(1):151–6. 44. Economic Research Service: www.ers.usda.gov. 45. Merkestein M, Brans MA, Luijendijk MC, de Jong JW, Egecioglu E, Dickson SL, et al. Ghrelin mediates anticipation to a palatable meal in rats. Obesity (Silver Spring). 2012;20(5):963–71. 46. Gimble JM, Sutton GM, Bunnell BA, Ptitsyn AA, Floyd ZE. Prospective influences of circadian clocks in adipose tissue and metabolism. Nat Rev Endocrinol. 2011;7(2):98–107. 2 The Pathophysiology of Obesity and Obesity-Related Disease 47. McAllister EJ, Dhurandhar NV, Keith SW, Aronne LJ, Barger J, Baskin M, et al. Ten putative contributors to the obesity epidemic. Crit Rev Food Sci Nutr. 2009;49(10):868–913. 48. Grant JL, MacKay KC, Manuel PM, McHugh TL. Barriers to optimizing investments in the built environment to reduce youth obesity: policy-maker perspectives. Can J Public Health. 2010;101(3):237–40. 49. Roseboom TJ, van der Meulen JH, Ravelli AC, Osmond C, Barker DJ, Bleker OP. Effects of prenatal exposure to the Dutch famine on adult disease in later life: an overview. Mol Cell Endocrinol. 2001;185(1–2):93–8. 50. Dabelea D, Hanson RL, Lindsay RS, Pettitt DJ, Imperatore G, Gabir MM, et al. Intrauterine exposure to diabetes conveys risks for type 2 diabetes and obesity: a study of discordant sibships. Diabetes. 2000;49(12):2208–11. 51. Shi H, Strader AD, Woods SC, Seeley RJ. The effect of fat removal on glucose tolerance is depot specific in male and female mice. Am J Physiol Endocrinol Metab. 2007;293(4):E1012–20. 52. Cypress AM, Lehman S, Williams G, Tal I, Rodman D, Goldfine AB, et al. Identification and importance of brown adipose tissue in adult humans. N Engl J Med. 2009;360:1509–17. 53. Pasarica M, Sereda OR, Redman LM, Albarado DC, Hymel DT, Roan LE, et al. Reduced adipose tissue oxygenation in human obesity: evidence for rarefaction, macrophage chemotaxis, and inflammation without an angiogenic response. Diabetes. 2009;58(3):718–25. 54. Ye J. Emerging role of adipose tissue hypoxia in obesity and insulin resistance. Int J Obes. 2009;33(1):54–66. 55. O’Rourke RW, White AE, Metcalf MD, Olivas AS, Mitra P, Larison WG, et al. Hypoxia-induced inflammatory cytokine secretion in human adipose tissue stromovascular cells. Diabetologia. 2011;54(6):1480–90. 56. Cancello R, Henegar C, Viguerie N, Taleb S, Poitou C, Rouault C, et al. Reduction of macrophage infiltration and chemoattractant gene expression changes in white adipose tissue of morbidly obese subjects after surgery-induced weight loss. Diabetes. 2005;54(8):2277–86. 57. Kamei N, Tobe K, Suzuki R, Ohsugi M, Watanabe T, Kubota N, et al. Overexpression of monocyte chemoattractant protein-1 in adipose tissues causes macrophage recruitment and insulin resistance. J Biol Chem. 2006;281(36):26602–14. 58. Weisberg SP, Hunter D, Huber R, Lemieux J, Slaymaker S, Vaddi K, et al. CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J Clin Invest. 2006;116(1):115–24. 59. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007;117:89–93. 60. Wentworth JM, Naselli G, Brown WA, Doyle L, Phipson B, Smyth GK, et al. Pro-inflammatory CD11c+CD206+ adipose tissue macrophages are associated with insulin resistance in human obesity. Diabetes. 2010;59(7):1648–56. 61. Ozcan U, Yilmaz E, Ozcan L, Furuhashi M, Vaillancourt E, Smith RO, et al. Chemical chaperones reduce ER stress and restore glucose homeostasis in a mouse model of type 2 diabetes. Science. 2006;313(5790):1137–40. 62. Mogensen M, Sahlin K, Fernstrom M, Glintborg D, Vind BF, Beck-Nielsen H, et al. Mitochondrial respiration is decreased in skeletal muscle of patients with type 2 diabetes. Diabetes. 2007;56(6):1592–9. 63. Patel C, Ghanim H, Ravishankar S, Sia CL, Viswanathan P, Mohanty P, et al. Prolonged reactive oxygen species generation and nuclear factor-kappaB activation after a high-fat, high-carbohydrate meal in the obese. J Clin Endocrinol Metab. 2007;92(11):4476–9. 64. Hoehn KL, Salmon AB, Hohnen-Behrens C, Turner N, Hoy AJ, Maghzal GJ, et al. Insulin resistance is a cellular antioxidant defense mechanism. Proc Natl Acad Sci U S A. 2009;106(42):17787–92. 35 65. Nielsen S, Guo Z, Johnson CM, Hensrud DD, Jensen MD. Splanchnic lipolysis in human obesity. J Clin Invest. 2004;113(11):1582–8. 66. Thaler JP, Choi SJ, Schwartz MW, Wisse BE. Hypothalamic inflammation and energy homeostasis: resolving the paradox. Front Neuroendocrinol. 2010;31(1):79–84. 67. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI, et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–31. 68. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56:1761–72. 69. Pappo I, Becovier H, Berry EM, Freund HR. Polymyxin B reduces cecal flora, TNF production and hepatic steatosis during total parenteral nutrition in the rat. J Surg Res. 1991;51:106–12. 70. Smith BW, Adams LA. Nonalcoholic fatty liver disease and diabetes mellitus: pathogenesis and treatment. Nat Rev Endocrinol. 2011;7:456–65. 71. Mohanty SR, Troy TN, Huo D, O’Brien BL, Jensen DM, Hart J. Influence of ethnicity on histological differences in non-alcoholic fatty liver disease. J Hepatol. 2009;50(4):797–804. 72. Cohen JC, Horton JD, Hobbs HH. Human fatty liver disease: old questions and new insights. Science. 2011;332(6037):1519–23. 73. Donnelly KL, Smith CI, Schwarzenberg SJ, Jessurun J, Boldt MD, Parks EJ. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J Clin Invest. 2005;115:1343–51. 74. Fabbrini E, Mohammed BS, Magkos F, Korenblat KM, Patterson BW, Klein S. Alterations in adipose tissue and hepatic lipid kinetics in obese men and women with nonalcoholic fatty liver disease. Gastroenterology. 2008;134:424–31. 75. Tilg H, Jalan R, Kaser A, Davies NA, Offner FA, Hodges SJ, et al. Anti-tumor necrosis factor-alpha monoclonal antibody therapy in severe alcoholic hepatitis. J Hepatol. 2003;38:419–25. 76. Hirosumi J, Tuncman G, Chang L, Görgün CZ, Uysal KT, Maeda K, et al. A central role for JNK in obesity and insulin resistance. Nature. 2002;420:333–6. 77. Tilg H, Moschen AR. Evolution of inflammation in nonalcoholic fatty liver disease: the multiple parallel hits hypothesis. Hepatology. 2010;52:1836–46. 78. Centers for Disease Control and Prevention: www.cdc.gov. 79. Juonala M, Magnussen CG, Berenson GS, Venn A, Burns TL, Sabin MA, et al. Childhood adiposity, adult adiposity, and cardiovascular risk factors. N Engl J Med. 2011;365(20):1876–85. 80. Hong EG, Ko HJ, Cho YR, Kim HJ, Ma Z, Yu TY, et al. Interleukin-10 prevents diet-induced insulin resistance by attenuating macrophage and cytokine response in skeletal muscle. Diabetes. 2009;58:2525–35. 81. Kim HJ, Higashimori T, Park SY, Choi H, Dong J, Kim YJ, et al. Differential effects of interleukin-6 and -10 on skeletal muscle and liver insulin action in vivo. Diabetes. 2004;53:1060–7. 82. Faghihimani E, Aminorroaya A, Rezvanian H, Adibi P, Ismail-Beigi F, Amini M. Salsalate improves glycemic control in patients with newly diagnosed type 2 diabetes. Acta Diabetol. 2011;50:537–43.. [Epub ahead of print] 83. Ursini F, Naty S, Grembiale RD. Infliximab and insulin resistance. Autoimmun Rev. 2010;9(8):536–9. 84. Larsen CM, Faulenbach M, Vaag A, Vølund A, Ehses JA, Seifert B, et al. Interleukin-1 receptor antagonist in type 2 diabetes mellitus. N Engl J Med. 2007;356:1517–26. 85. Cruz AB, Amatuzio DS, Grande F, Hay LJ. Effect of intraarterial insulin on tissue cholesterol and fatty acids in alloxan-diabetic dogs. Circ Res. 1961;9:39–43. 86. Bugiardini R, Manfrini O, Pizzi C, Fontana F, Morgagni G. Endothelial function predicts future development of coronary artery disease: a study of women with chest pain and normal coronary angiogram. Circulation. 2004;109:2518–23. 36 87. Charo IF, Taub R. Anti-inflammatory therapeutics for the treatment of atherosclerosis. Nat Rev-Drug Discov. 2011;10:365–76. 88. Singh SK, Grifson JJ, Mavuduru RS, Agarwal MM, Mandal AK, Jha V. Serum leptin: a marker of prostate cancer irrespective of obesity. Cancer Biomark. 2010;7(1):11–5. 89. Baglietto L, English DR, Hopper JL, Morris HA, Tilley WD, Giles GG. Circulating insulin-like growth factor-I and binding protein-3 R. W. O’Rourke and the risk of breast cancer. Cancer Epidemiol Biomark Prev. 2007;16(4):763–8. 90. Manning BD. Balancing Akt with S6K: implications for both metabolic diseases and tumorigenesis. J Cell Biol. 2004;167(3):399–403. 91. Decensi A, Puntoni M, Goodwin P, Cazzaniga M, Gennari A, Bonanni B, et al. Metformin and cancer risk in diabetic patients: a systematic review and meta-analysis. Cancer Prev Res (Phila). 2010;3(11):1451–61. 3 History of the Development of Metabolic/Bariatric Surgery Elias Chousleb, Jaime A. Rodriguez, and J. Patrick O’Leary Chapter Objectives 1. Review the history and evolution of bariatric and metabolic surgery. 2. Learn from previous experiences in the development of bariatric and metabolic surgeries in order to continue to improve safety and efficacy. History of Bariatric Surgery The odyssey of the surgical intervention for the treatment of serious obesity began as all odysseys begin when a problem was recognized and a prepared mind coupled divergent observations and asked the question, “Why not?” In this case, the problem was severe incapacitating obesity, and the observation was that when individuals underwent resection of the greater portion of their small intestine, weight loss ensued even if the individual was of normal weight at the outset. As the world came out of the Second World War, farming in many areas became more mechanized, manpower became available, foodstuffs became extremely affordable, and the fast-food industry emerged. The US agriculture thrived as did the urbanization of not only the United States but also the world. Preservatives were added to extend the shelf life of food. The prevalence of obesity skyrocketed. At this time young physicians who had their medical education interrupted by the call to military duty returned to complete their specialty training. Several institutions, notably the University of Minnesota, shunted a substantial part of E. Chousleb · J. A. Rodriguez · J. P. O’Leary (*) Department of Surgery, Florida International University, Herbert Wertheim College of Medicine, Miami, FL, USA e-mail: olearyp@fiu.edu this work force into research laboratories, many of which were committed to unraveling the mystery of the gastrointestinal tract. Working in the environment nurtured by Owen H. Wangensteen and under the direct tutelage of Richard L. Varco, one such individual, John Linner, set about transposing segments of the small intestine to better understand the physiologic role of the jejunum as compared to the ileum. The job was arduous, the studies sophisticated, and the research laboratory was not air-conditioned. The studies were done in a canine model and were of such quality that the work was selected for presentation at the American Surgical Spring Meeting in 1954 [1]. As a part of the presentation, a comment was made about a young, seriously obese woman with heart disease that had undergone an operation to bypass the majority of her small intestine. She had lost weight, and her cardiac disease had stabilized. In the discussion of this paper, Philip Sandblom from Sweden commented that a Swedish surgeon, Viktor Henriksson, had performed a similar procedure in a small number of patients, and although they had experienced “some difficult situations of nutritional balance,” they had experienced weight loss. The patient described by Linner underwent a revision of the primary bariatric procedure in 1981 and survived to age 61 when she died of a cardiac event (Linner JG, 1985, personal communication). Although still heavy, she had not regained her original severely obese state. John Kral researched the patients operated upon by Henriksson and found they also experienced long-term control of their obesity (Kral J, 1985, personal communication). Based in part on these results, Payne (a surgeon), DeWind (a gastroenterologist), and Commons (a pathologist) as part of a large study on “morbid” obesity (“morbid” a term coined by Payne to encourage insurance companies to pay for these procedures) performed an end-to-side jejunocolic shunt in ten patients [2] (Fig. 3.1). This was a part of an experimental study in which patients consented to a large number of baseline studies and to similar follow-up studies to characterize the effects that the procedure produced. The results of these studies were published in great detail and in a style that was © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_3 37 38 E. Chousleb et al. Fig. 3.1 Jejunocolic bypass Fig. 3.2 End-to-side jejunoileal bypass popular at that time. Weight loss occurred in each of the patients. One patient died 6 months after the procedure of a pulmonary embolism. She had an antecedent history of pulmonary emboli. The protocol included the reestablishment of continuity of the gastrointestinal tract when optimal weight had been achieved. In the six patients in whom continuity of the gastrointestinal tract was restored, all regained their previous obese state. The three remaining patients had their jejunocolic shunt revised to an end-to-side jejunoileal shunt (Fig. 3.2). One of these patients, in which a substantial amount of jejunum was placed back in continuity, experienced weight regain to her preoperative level. The authors observed that if a reasonable amount of jejunum (14 in.) and a smaller portion of ileum (4 in.) were left in continuity with the ingested food, weight loss could be maintained. In a later publication, Payne and DeWind reported acceptable weight loss results in a large number of patients [3]. By the late 1960s and early 1970s, there were a number of reports of successful weight loss being produced by jejunoileal or jejunocolic shunts. Each of these studies identified complications that were directly associated with the intestinal bypass procedure. As these reports became more numerous, it produced an extremely fertile field for a number of researchers to investigate the various mechanisms of action that produced the metabolic aberration. Perhaps, the most interesting and best studied side effect of intestinal shunting was the development of liver failure in certain patients. In Payne’s original report, all of the patients had liver biopsies, and the vast majority demonstrated steatosis of the liver. Many surgeons paid little heed to this observation. After all, the patient was fat and had fat everywhere else, why not in the liver. If the excluded limb of the intestine was resected, liver failure did not occur, although these patients did experience malabsorption and weight loss. When liver failure 3 History of the Development of Metabolic/Bariatric Surgery 39 occurred, biopsies of the liver looked identical to alcohol-­ Bariatric surgery was often looked upon as a nuisance that induced cirrhosis, even to the point of including Mallory’s had a tendency to congest intensive care units with long-term bodies. Many curmudgeons opined that these patients must stay patients. Academic advancement for young surgeons be closet alcoholics. That was clearly not the case. Certain occurred in spite of their involvement in bariatric surgery not investigators demonstrated bacterial overgrowth of gram-­ because of their achievements in this area. National meetings negatives and anaerobic bacteria in the excluded limb of the rarely accepted papers about bariatric surgery, and when intestine along with morphological changes in the intestinal papers were accepted, they almost always dealt with the wall (separation of tight junctions between enterocytes) and management of complications that might be pertinent to simfelt that this played a role [4]. These investigators coined the ilar complications that occurred in normal weight individuterm “enterohepatic syndrome.” Other complications of als. The papers were always positioned at the worst spot on intestinal shunting procedures included malnutrition, vita- the program. min deficiencies (especially of fat-soluble vitamins), electroBy the early 1970s, certain surgeons became sensitized to lyte abnormalities (especially of divalent cations), ketosis, the complications associated with malabsorptive procedures iron malabsorption, hyperoxaluria, nephrolithiasis, migra- and looked for alternatives. From a simplistic standpoint, if tory arthralgia, profound inflammation of synovial lined allowing patients to eat large volumes of food and then interspaces, and in some individuals weight regain. Although the rupting absorption by short-circuiting the intestine did not benefits were profound, so were the complications. The work, perhaps limiting intake would. second-­ever National Institutes of Health (NIH) Consensus At the University of Iowa, there were strong connections Development Conference was held in 1978 [5]. The focus to the University of Minnesota. Not only were they in relative was the treatment of morbid obesity, including surgical inter- proximity, but many of the faculty at Iowa had been educated vention. Although the recommendations were favorable for in part at the University of Minnesota. Edward E. Mason was certain operative procedures in the treatment algorithm, it interested in the gastrointestinal tract and specifically in pepwas felt that the risk-benefit ratio for intestinal shunting pro- tic ulcer disease. Working with Chikashi “Chick” Ito, Mason cedures was too high to recommend routine use. performed a side-to-side anastomosis between the very upper Up to this time, the academic surgical community had third of the divided stomach and a loop of the jejunum to shown little interest in the development of bariatric surgery. treat duodenal ulcers disease (Fig. 3.3). A number of patients The individuals doing bariatric surgery were felt to be some- were obese, and Mason observed that although the procedure what of a renegade group of surgeons who were involved in did not control the ulcers, it was associated with weight loss. the treatment of a “condition” that was simply the end result He reported this in a 1967 publication during the peak of of gluttony and sloth. This prejudiced view augmented the discrimination against the patients, their disease, and the surgeons and staff that treated them. It was a commonly held belief that seriously obese individuals simply lacked willpower. Surgeons who lowered themselves to treat individuals that were affected by obesity, which was not a serious disease (like cancer), deserved the headaches inherent in the care of such patients. It was a waste of resources that could have been used in the treatment of “real” surgical problems such as duodenal ulcer disease. There was little or no appreciation that obesity was a disease and intimately associated with other diseases such as type II diabetes, hypertension, cardiovascular disease, and an increased incident of certain malignancies. There was a failure to understand that the underlying inflammatory process associated with severe obesity affected every system in the body to a greater or lesser degree. It was not realized that control of the obesity would be associated with increased longevity and general health. There was a real schism between believers that obesity was a disease state and those that did not adhere to this premise. Department chairs grudgingly allowed young surgical faculty to do bariatric surgery as it produced volume in the operating room and technical experience for residents. Fig. 3.3 Side-to-side anastomosis 40 Fig. 3.4 Roux-en-Y limb for gastrojejunostomy popularity for the jejunoileal bypass [6]. The paper was somewhat confusing to the surgical community as it reported results for patients with two different diseases. As these patients were followed longer term, weight regain was observed. Over the subsequent 8 years, Mason published three more papers modifying the procedure by first making the anastomosis between the stomach and the jejunum smaller and then substantially decreasing the gastric pouch size. The problems were multiple. The procedure was performed high in the abdomen and therefore was technically demanding, and the enlarged left lobe of the liver was often problematic. Staplers were not available in the early years. Although early weight loss was obtained, weight regain occurred in many patients by 6 months. Because the procedure involved a loop of jejunum, regurgitation of small bowel content into the gastric pouch frequently occurred. Alden solved some of the problems by doing away with the loop and creating in Roux-en-Y limb for the gastrojejunostomy [7] (Fig. 3.4). Mason continued to modify his procedure and by the mid-­ 1970s had first performed a gastric partition with the opening on the greater curvature of the stomach. This was done only in a small number of patients and did not seem to produce long-term weight loss (Fig. 3.5). By 1975, Tretbar, Echout, Fabito, Laws, O’Leary, and others had performed a vertical gastric partition along the lesser curvature of the stomach and controlled the outlet using a variety of devices from chromic suture to a silicone ring [8] (Fig. 3.6). Staplers were critical in the development of such a partition. Mason modified his approach by placing an end-to-end anastomosis E. Chousleb et al. Fig. 3.5 Gastric partition with the opening on the greater curvature of the stomach Fig. 3.6 Vertical gastric partition along the lesser curvature of the stomach with the outlet controlled with a silicone ring (EEA) stapler through the stomach at the distal end of the lesser curvature cylindrical gastric tube and placing a piece of Marlex® mesh through the hole and back up through an aperture at the lesser curvature of the stomach sparing the nerve of Latarjet [9] (Fig. 3.7). This vertical banded gastro- 3 History of the Development of Metabolic/Bariatric Surgery Fig. 3.7 End-to-end anastomosis through the stomach at the distal end of the lesser curvature cylindrical gastric tube with a piece of Marlex mesh through the hole and back up through an aperture at the lesser curvature of the stomach plasty gained considerable popularity and for a period of time in the early 1980s was probably the most commonly performed bariatric operation in the United States. The same NIH consensus conference that spelled the demise of the jejunoileal bypass surgery opened an alternate avenue for gastric restrictive procedures. A variety of different mechanisms had been explored to partition the stomach. Various surgeon pioneers began to perfect the operative techniques surrounding gastric bypass procedures. Investigators from the University of Kentucky and the University of North Carolina published comparison studies between the intestinal bypass procedure and gastric bypass [10, 11]. Complications were clearly less in the gastric procedures, and weight loss was equivalent. During the 1980s, Mason continued to champion gastric restriction using the banded gastroplasty. Various modifications of these procedures were proposed. In an ingenious modification of the silastic ring championed by Laws, L. Kuzmak invented a silastic ring with a small balloon embedded on the inner aspect of the ring that could be accessed from a subcutaneously placed reservoir [12]. This allowed calibration of the outflow lumen (Fig. 3.8). With this innovation, adjustable gastric banding was born. At this phase in the development of bariatric surgery, which occurred in the 1980s, a number of individuals adopted the philosophy that gastric restrictive procedures—including gastric bypass, which was thought at that time to be primarily a restrictive procedure—were associated with less in the 41 Fig. 3.8 Adjustable gastric banding. A silastic ring with a small balloon embedded on the inner aspect of the ring that could be accessed from a subcutaneously placed reservoir way of postoperative complications, produced satisfactory weight loss, were associated with amelioration of the complications of obesity, and were technically reasonable to perform. Gastric restrictive procedures benefited enormously from advances in technology, especially in the area of stapling devices. Surgeons also learned many invaluable lessons as to the management of obese individuals after other intra-abdominal procedures. These lessons were accepted by the general surgeons, who were not necessarily doing bariatric surgery but operating on seriously obese people for other causes. Not all of the advances in bariatric surgery were confined to the North American continent. Bariatric surgery was beginning to become recognized in Europe, South and Central America, and to a lesser degree Asia. One of the most prolific writers from the European continent was Nicola Scopinaro, who in 1979 had devised an operation he termed the biliopancreatic diversion (BPD) [13]. Scopinaro performed a generous gastrectomy, usually leaving a gastric remnant about one-third the size of the original stomach. He then divided the small intestine at about its midpoint and brought the ileal end up to be anastomosed to the stomach remnant. The other end of the intestine that carried the biliary and pancreatic excretions was anastomosed to the side of the ileum, approximately 120 cm from the ileocecal valve (Fig. 3.9). This produced an abbreviated channel where the digestive juices mixed with ingested food. Scopinaro reported excellent weight loss results. His patients underwent a large number of metabolic studies that demonstrated amelioration of many of the comorbidities associated with morbid obesity. 42 E. Chousleb et al. detailed physiology was at a much higher level than ever before, although still quite incomplete. The vast majority of procedures were gastric restrictive procedures, with the most common operation performed being the gastric bypass. The bariatric surgical community could now perform this type of surgical intervention with an operative mortality of less than 1% and morbid complications occurring in less than 6% of patients. The beneficial effects had now been clearly documented in certain areas. Although insurance coverage was not widely available, certain payers were beginning to cover the procedures. The field of surgery for morbid obesity was poised at the precipice, awaiting the next great breakthrough. These change agents came in two forms. The first was a report by MacDonald and Pories published in 1995 detailing the beneficial effect seen in obese individuals with adult-onset (type II) diabetes who had undergone a Roux-en-Y gastric bypass [15]. It was known that diabetes was ameliorated by weight loss. Previous studies, in the early 1980s, dealt with changes in insulin resistance and glucose metabolism after intestinal shunting procedures [16]. Insulin resistance improved, and hyperglycemia disappeared even before weight loss had occurred. This report went virtually unnoticed, in part because intestinal shunting procedures were out of favor. The Pories report about glucose metabolism and the amelioration of diabetes was well received in the bariatric surgical community but less well received in the general medical community. The report showed that insulin levels plummeted, while glucose metabolism improved, suggesting a change in insulin resistance. Eight years later, in 2003, Schauer reported similar results Fig. 3.9 A generous gastrectomy leaving a gastric remnant about one-­ third the size of the original stomach. The small intestine is divided at in a large cohort of patients who had either impaired testing about its midpoint, and the ileal end is brought up to be anastomosed to glucose levels or type 2 diabetes mellitus (T2DM) [17]. the stomach remnant. The other end of the intestine that carried the bili- Shortly thereafter, prompted by these observations, a summit ary and pancreatic excretions is anastomosed to the side of the ileum, was convened in Rome by a diverse group of individuals approximately 120 cm from the ileocecal valve with an interest in T2DM. This summit was well attended by a large number of scientific organizations. A consensus was He did acknowledge many of the side effects seen in the pre- reached and published in 2010 [18]. viously discussed malabsorptive procedures, especially those Perhaps the most important results of this meeting were having to do with iron and divalent cation absorption as well the creation of a research agenda and the more widespread as absorption of fat-soluble vitamins. From the literature, it understanding of some of the mechanisms by which diabetes would appear that his patients required diligent and pro- was controlled with surgical intervention. It was understood longed follow-up, but with this type of management, that something was happening to these patients that went far Scopinaro reported excellent long-term results [14]. beyond simply rerouting the food flow in the intestines— something metabolic. Although Wolfe had introduced the term “metabolic intestinal surgery” in the mid-1970s, it was Metabolic only now as surgeons begin to truly appreciate the magnitude of what was being accomplished that the name “metabolic” As the decade of the 1980s drew to a close, the field of bar- resurfaced [19]. iatric surgery had stabilized. Much had been learned and The third NIH consensus conference on obesity, in 1991, applied over the preceding 25 years. The surgeon’s knowl- found that surgical intervention of morbid obesity amelioedge about the disease had improved substantially. The rated many of the comorbidities associated with obesity. In understanding of complex hormonal mechanisms and 2008, the membership of the American Society of Bariatric 3 History of the Development of Metabolic/Bariatric Surgery Surgery elected at the annual business meeting to change the name of the society by adding “Metabolic” to the organization’s title. This seminal change refocused efforts on understanding how these procedures worked. In some ways, this validated earlier bariatric surgeons who had demanded that patients be followed long term and that their results be published using defined parameters of success. The conference went on to conclude that the risk-benefit ratio in certain patients favored surgical intervention [20]. Minimally Invasive Techniques On the second front, in the early 1990s, there was a pivotal technical revolution in general surgery. Surgeons had begun to explore the laparoscopic approach to certain general surgical procedures. Obstetrics and gynecology physicians had known for years that the abdominal cavity could be approached by minimally invasive techniques. These techniques were usually limited to observation and, perhaps through biopsy, diagnosis. Therapeutic interventions had not been a part of their armamentarium. Advances in digital imaging, light sources, miniaturization of cameras, and advanced instrumentation would allow therapeutic intervention to become possible. With these advances, minimally invasive surgery was spawned. As laparoscopic cholecystectomy became commonplace, the tsunami of minimally invasive operations followed. The first minimally invasive gastric restrictive procedure was not far behind. The first laparoscopic gastric bypass operation in the United States was performed by Wittgrove and Clark in October 1993, using a retrocolic limb with a circular stapler anastomosis for the gastrojejunostomy [21]. Just as in the open procedure, the three key components to the creation of an effective gastric bypass were the creation of a small gastric pouch, a restrictive gastrojejunal anastomosis, and the creation of a roux limb to promote malabsorption. Initially technical constraints made this operation very difficult, but as experience with laparoscopic surgery grew and as suturing, stapling, and electrocoagulation devices became readily available, the popularity of laparoscopic gastric bypass increased rapidly. By the late 1990s almost every academic department of surgery had created a division that performed minimally invasive bariatric procedures. The era of minimally invasive surgery revolutionized the surgical management of obesity. The number of bariatric procedures multiplied exponentially allowing surgeons to care for patients in a much safer and effective fashion. Laparoscopy allowed surgeons to perform these complex gastrointestinal operations with a level of safety that has never before been seen. This improved morbidity and mortality profile made bariatric surgery increasingly more attractive to patients, surgeons, and referring physicians alike. 43 The landscape had truly changed – where operative mortality had been less than 1%, now the operative mortality plummeted to less than 0.2%. The rate of complications fell to a third of complication rate for open procedures [22]. Hospital stays went from 3 to 5 days down to 2 days. The use of laparoscopy in bariatric surgery resulted in reduced impairment of pulmonary function [23], less intraoperative blood loss, shortened length of hospital stay, decreased rates of wound infection, and incisional hernias becoming rare [24]. Reduction in complication rates resulted in decreased costs. Patients routinely returned to work in less than 2 weeks. There was another innovation that affected the safety of surgery for obesity and related disease. The first successful laparoscopic banding procedure was published in 1993 by Broadbent [25]. The authors reported the laparoscopic placement of a nonadjustable gastric band in a 16-year-old female the year prior. Catona also published a series of patients who underwent nonadjustable gastric banding using a laparoscopic approach in the same time frame [26]. These procedures were similar to the Laws procedure without the lesser curvature partition. During the same time, Belachew designed an adjustable gastric band that could be placed using laparoscopic techniques. He described this procedure in a porcine model [27]. This was similar to the band patented by Kuzmak a decade earlier. A large number of bands were placed in Australia, Latin America, and Europe prior to approval of the device in the United States. Subsequently, O’Brien from Australia published a review of laparoscopic adjustable gastric banding that showed durable weight loss of 47% excess body weight (EBW) in patients followed up to 15 years [28]. In 1986, Hess and Hess performed an open procedure adding a sleeve gastrectomy to the original biliopancreatic bypass procedure and modified the anastomosis to a duodenojejunal (BPD-DS) configuration (Fig. 3.10) [29]. Some of the advantages of BPD-DS included the avoidance of dumping syndrome and marginal ulceration by preserving the innovated pylorus and creating the anastomosis between the duodenum and the jejunum. A common channel of 100 cm was created; therefore, malabsorption of protein and calories were increased as compared to Roux-en-Y gastric bypass. The first laparoscopic duodenal switch was performed by Ren and Gagner in 1999, as a modification of the original Scopinaro procedure [30]. Laparoscopic BPD-DS has been associated with larger weight loss when compared to all other bariatric procedures. It was reserved for patients with a body mass index (BMI) of 60 or greater, but due to technical difficulty as well as concerns for nutritional deficiency, the procedure has not been widely adopted in the United States. Throughout the history of bariatric surgery, there has been avid interest in developing safer procedures with less complications and equivalent effectiveness in the short and long 44 E. Chousleb et al. Fig. 3.10 An open procedure adding a sleeve gastrectomy to the original biliopancreatic bypass procedure and modifying the anastomosis to a duodenojejunal (BPD-DS) configuration Fig. 3.11 Sleeve gastrectomy term. Johnston, in 1987, performed the first Magenstrasse and Mill procedure [31]. The idea was to create a safe simple alternative to the gastric bypass and the vertical banded gastroplasty. The Magenstrasse referred to a thin tube created from the lesser curvature of the stomach and the Mill referred to the antrum. The Magenstrasse and Mill procedure was created by using a circular stapler to create a defect in the antrum and then creating a narrow tube along the lesser curvature initially over a 40 Fr bougie. The diameter of the bougie was then reduced to a 32 Fr to optimize weight loss. The technique was subsequently improved by simply resecting the greater curvature of the stomach. The result was the sleeve gastrectomy, which had previously been performed as part of the restrictive component of the duodenal switch by Hess and then Marceau [32]. Sleeve gastrectomy was initially used as part of a two-­ step procedure in high risk (BMI > 60) patients (Fig. 3.11). Close follow-up of these patients revealed substantial weight loss and resolution of comorbidities with the sleeve gastrectomy alone [33]. The indications for laparoscopic sleeve gastrectomy (LSG) as a stand-alone procedure were published in 2008 [34]. The popularity of the LSG has grown dramatically due to a perceived simplicity of the surgical technique and adequate resolution of comorbidities. However, long-­term studies on effectiveness and difficult to treat complications, such as leak, require further evaluation. Hormonal Weight Loss As a better understanding of gut peptides emerged, innovative procedures were developed to produce hormonally induced weight loss. One such procedure targeted the gut neurohormonal activity by producing an ileal interposition in upper intestines. Harkening back to the early work of Linner, Mason in 1999 interposed the ileum to a position in the proximal jejunum and produced weight loss [35]. A 170–200-cm-­long portion of ileum was isolated leaving 30 cm of distal ileum. This segment was relocated 50 cm distal to the ligament of 3 History of the Development of Metabolic/Bariatric Surgery 45 procedures. It seemed the complication rate was unacceptably high. The underlying bias and discrimination against patients affected by obesity continued to create a public environment of blame for both the patient and the surgeons who operated on them. The ASMBS moved not only to create educational programs but also to establish accreditation for surgeons and institutions that were performing bariatric surgical intervention. Conclusion Fig. 3.12 The ileum is interposed to a position in the proximal jejunum. A 170–200-cm-long portion of the ileum is isolated leaving 30 cm of the distal ileum. This segment is relocated 50 cm distal to the ligament of Treitz utilizing two jejunoileal anastomoses. Bowel continuity is restored with a third jejunoileal anastomosis Treitz utilizing two jejunoileal anastomoses. Bowel continuity was restored with a third jejunoileal anastomosis (Fig. 3.12). Multiple peptides have been studied, some of which surely play a role in food intake and satiety. The most fertile area of current research is in establishing the actual mechanism of action by which metabolic procedures work and relating that to the pathophysiology of obesity and related disorders. It is through this path of investigation that future innovation will improve the safety and effectiveness of these procedures. Accreditation In 2004, an impending crisis loomed that centered in some ways around ethics. Two separate factors contributed to the conundrum; laparoscopic procedures were now performed commonly by a markedly increased number of surgeons who felt comfortable approaching the abdomen using a minimally invasive approach. At the same time, more insurance coverage was available for patients needing surgical interventions for their obesity. Although the operative procedure itself was becoming better understood from a technical standpoint, many surgeons, with only minimal skills, were operating on patients who were at high risk. A few high-profile complications attracted attention both on television and in print media, with challenging questions being asked about the level of training of bariatric surgeons and about the necessity of these At the close of the first decade of the twenty-first century, several things would appear to be unequivocally true. Firstly, surgical intervention in the treatment of morbid obesity has an acceptable risk-benefit ratio and produces amelioration or control of many of the diseases associated with serious obesity. Secondly, the minimally invasive approach has contributed remarkably to the improvement in the risk-benefit ratio. Thirdly, although much remains to be understood, metabolic/ bariatric surgery has emerged from the shadows of charlatanism into the mainstream of general surgery. The baby was not thrown out with the bathwater and has emerged on the surgical stage as a vibrant, healthy adolescent. References 1. Kremen AJ, Linner JH, Nelson CH. An experimental evaluation of the nutritional importance of proximal and distal small intestine. Ann Surg. 1954;140:439. 2. Payne JH, LT DW, Commons RR. Metabolic observations in patients with jejunocolic shunts. Am J Surg. 1963;106:273. 3. Payne JH, LT DW. Surgical treatment of obesity. Am J Surg. 1969;118:141. 4. O’Leary JP. Historical perspective on intestinal bypass procedures. In: Griffen WO, Printen KJ, editors. Surgical management of morbid obesity. New York: Marcel Dekker; 1987. p. 1–26. 5. Surgical Treatment of Morbid Obesity. NIH consensus statement 1978;1(10):39–41. 6. Mason EE, Ito C. Gastric bypass in obesity. Surg Clin North Am. 1967;47:1345. 7. Alden JF. Gastric and jejunoileal bypass. Arch Surg. 1977;112:799–806. 8. O’Leary JP. Partition of the lesser curvature of the stomach in morbid obesity. Surg Gynecol Obstet. 1982;154:85. 9. Mason EE, Printen KJ, Bloomers TJ, Scott DH. Gastric bypass for obesity after ten years experience. Int J Obes. 1978;2:197–205. 10. Buckwalter JA. A prospective comparison of the jejunoileal and gastric bypass operations for morbid obesity. World J Surg. 1977;1:757. 11. Griffen WO, Young VL, Stevenson CC. A prospective comparison of gastric and jejunoileal procedures for morbid obesity. Ann Surg. 1977;186:500. 12. Kuzmak L. Silicone gastric banding: a simple and effective operation for morbid obesity. Contemp Surg. 1986;28:13–8. 13. Scopinaro N, Gianetta E, Civalleri D, Bonalumi U, Bachi V. Bilio-­ pancreatic bypass to obesity: II. Initial experience in man. Br J Surg. 1979;55:518. 46 14. Scopinaro N, Adam GF, Marinari GM. Biliopancreatic diversion. World J Surg. 1998;22:936–46. 15. Pories WJ, Swanson MS, KG MD, Long SB, Morris PG, Brown BM, et al. Who would have thought it? An operation proves to be the most effective therapy for adult onset diabetes mellitus. Ann Surg. 1995;222(3):339–52. 16. O’Leary JP, Duerson MC. Changes in glucose metabolism after jejunoileal bypass. Surg Forum. 1980;31:87. 17. Schauer PR, Burguera B, Ikramuddin S, Cottam D, Gourash W, Hamad G, et al. Effect of laparoscopic Roux-en-Y gastric bypass on type 2 diabetes mellitus. Ann Surg. 2003;238(4):467–85. 18. Rubino F, Kaplan LM, Schauer PR, Cummings DE. Diabetes surgery summit delegates. The diabetes surgery summit consensus conference: recommendations for the evaluation and use of gastrointestinal surgery to treat type 2 diabetes mellitus. Ann Surg. 2010;251(3):399–405. 19. Starkloff GB, Donovan JF, Ramach R, Wolfe BM. Metabolic intestinal surgery its complications and management. Arch Surg. 1975;110:652–7. 20. Gastrointestinal Surgery for Severe Obesity. NIH consensus statement 1991;9(1):1–20. 21. Wittgrove AC, Clark GW, Tremblay LJ. Laparoscopic gastric bypass, Roux en Y: preliminary report of five cases. Obes Surg. 1994;4(4):353–7. 22. Miller MR, Choban PS. Surgical management of obesity: current state of procedure evolution and strategies to optimize outcomes. Nutr Clin Pract. 2011;26(5):526–33. 23. Nguyen NT, Lee SL, Goldman C, Fleming N, Arango A, McFall R, et al. Comparisons of pulmonary function and postoperative pain after laparoscopic versus open gastric bypass: a randomized trial. J Am Coll Surg. 2001;192(4):469–77. 24. Nguyen NT, Hinojosa M, Fayad C, Varela E, Wilson SE. Use and outcomes of laparoscopic versus open gastric bypass at academic medical centers. J Am Coll Surg. 2007;205(2):248–55. E. Chousleb et al. 25. Broadbent R, Tracey M, Harrington P. Laparoscopic gastric banding: a preliminary report. Obes Surg. 1993;3:63. 26. Catona A, Gossenberg M, La Manna A, Mussini G. Laparoscopic gastric banding: preliminary series. Obes Surg. 1993;3:207. 27. Belachew M, Legrand MJ, Defecherux TH, Burtheret MP, Jacquet N. Laparoscopic adjustable silicone gastric banding in the treatment of morbid obesity. A preliminary report. Surg Endosc. 1994;8:1354–6. 28. O’Brien PE, Mac Donald L, Anderson M, Brennan L, Brown WA. Long-term outcomes after bariatric surgery. Fifteen year follow up of adjustable gastric banding and a systematic review of the bariatric surgical literature. Ann Surg. 2013;257(1):87–94. 29. Hess DS, Hess DW. Biliopancreatic diversion with a duodenal switch. Obes Surg. 1998;8:267–82. 30. Ren CJ, Patterson E, Gagner M. Early results of laparoscopic biliopancreatic diversion with duodenal switch: a case series of 40 consecutive patients. Obes Surg. 2000;10(6):514–23. 31. Johnston D, Dachtler J, Sue-Ling HM, King RF, Martin G. The magenstrasse and mill operation for morbid obesity. Obes Surg. 2003;13(1):10–6. 32. Marceau P, Biron S, Bourque RA, Potvin M, Hould FS, Simard S. Biliopancreatic diversion with a new type of gastrectomy. Obes Surg. 1993;3(1):29–35. 33. Rosenthal RJ. International sleeve gastrectomy expert panel consensus statement: best practice guidelines based on >12,000 cases. Surg Obes Relat Dis. 2012;8:8–19. 34. Talebpour M, Amoli BS. Laparoscopic total gastric placation in morbid obesity. J Laparoendosc Adv Surg Tech A. 2007;17(6):793–8. 35. Mason EE. Ileal (correction of ilial) transportation and enteroglucagon/GLP-1 in obesity (and diabetic?) surgery. Obes Surg. 1999;9(3):223–8. 4 The History of the American Society for Metabolic and Bariatric Surgery Robin P. Blackstone Chapter Objectives 1. Review the history and evolution of the American Society for Metabolic and Bariatric Surgery (ASMBS). 2. Provide an overview of current programs and initiatives undertaken by the society. 3. Demonstrate the vision and leadership that led to the current state of metabolic and bariatric surgery within the United States. Introduction The history of the American Society for Metabolic and Bariatric Surgery (ASMBS) parallels the development of metabolic and bariatric surgery (MBS) closely. In studying this history, three distinct periods emerge. The first, the Era of Inquiry (1967–1988), established the initial scientific foundation of MBS. The second is the Era of Rapid Growth (1989–2004) during which the society supervised rapid growth in the number of surgeons and programs and the growth of integrated multidisciplinary teams. The number of cases grew rapidly spurred on by laparoscopic access and with increasing diversity in procedures performed and use of devices. The third period, the Era of Quality and Engagement (2004 to present), established the society’s focus on safety and increasing engagement with other stakeholders. These stakeholders include medical colleagues, international surgeons, the American College of Surgeons, medical societies dedicated to the management of obesity, and patient advocacy groups. Together we have focused on building an infrastructure for the population management of obesity in the United States and globally R. P. Blackstone (*) Metabolic and Bariatric Surgery, Banner University Medical Center – Phoenix, Phoenix, AZ, USA e-mail: robin.blackstone@bannerhealth.com using a similar collaborative model that exits between surgery and medicine in other disciplines. Most patients with obesity are treated in a community hospital setting. The participation of surgeons and their integrated health partners in using high-quality data for ongoing quality improvement provides leadership to improve patient safety and experience of care within the community. The ASMBS has been successful in creating a high-value data-driven network of nationally accredited programs accepted as the foundation of quality by payers. Over the last interval, the network has been leveraged to address ongoing quality issues for underperforming centers focused on readmissions and enhanced recovery protocols. These efforts are transformational. MBS is conducted in the United States within a learning and continuously improving culture. It is the signal achievement of this era. The Era of Inquiry 1967–1988 From the beginning, MBS developed through multidisciplinary collaboration with the focus located at the University of Iowa (UI). At UI, a unique environment was created by the collaboration of surgeons, physiologists, biochemists, and integrated health professionals at the National Institutes of Health (NIH) General Clinical Research Center (GCRC). This Center, led by Edward Mason, MD (Fig. 4.1), presented its multidisciplinary findings in collaborative discussions of each other’s work. In 1967 in a symposium honoring Owen H. Wangensteen, MD, Dr. Mason’s mentor, Dr. Mason, and Chikashi Ito, PhD, presented the first case of gastric bypass. These meetings became more formal and became known as the Mason Surgical Treatment of Obesity Symposium Workshop in 1976, attracting national and international scientists and surgeons interested in treating the disease of obesity. With NIH funding, the group continued to explore the physiological and metabolic effect of gastric bypass and presented their findings at the American Surgical Association Meeting in 1969. © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_4 47 48 Fig. 4.1 Edward Mason, MD R. P. Blackstone 4 The History of the American Society for Metabolic and Bariatric Surgery Edward Mason writes about those days: The postgraduate course was started because of the increasing number of surgeons performing obesity surgery, who were communicating and sharing experiences and ideas by phone. Nicola Scopinaro, MD, from Genoa and surgeons from Sweden were early attendees. In 1977, there were 28 presentations and symposia listed for the meeting, which was held April 28 and 29 and called the Gastric Bypass Workshop. I found a copy of the bound paperback of 187 pages recording transcript of the 1977 meeting, which was distributed after the meeting. The last article is about plans for a Gastric Bypass Registry. The Workshop transcript was distributed a month after the meeting. It includes the presentations and discussion that were recorded and transcribed. Long-term results and prospective and larger trials began to contribute to the knowledge base, culminating in the first NIH consensus conference on December 4, 1978. This conference was of pivotal importance. It was at this conference that the jejunal-ileal bypass was shown to have substantial problems, and restriction (gastric bypass and vertical banded gastroplasty) was established as a credible procedure. The society was formed on this strong scientific foundation. John Kral writes: Having attended the Iowa City colloquia, the academic surgeons J D Halverson, J P O’Leary, H J Sugerman and myself, at the colloquia in 1983, met in a pub during the lunch break to propose expanding the colloquia to the format of scientific meetings with membership, program committees, minutes, abstract selection and “democratic” principles. That afternoon, June 3, 1983 at an impromptu business meeting of the attendees a proposal for the formation of a society for the study of obesity surgery was made and accepted. The aims of the society were to “develop guidelines for patient care, promote research into the outcomes and quality of bariatric surgeries and encourage an exchange of ideas among researchers and surgeons” [1]. In deference to Mason, the term “bariatric”—a continuation of his tradition of the colloquia—was adopted. In 1984, the first annual meeting of the American Society for Bariatric Surgery (ASBS) was held in Iowa City. The meeting attracted more than 150 participants and 36 oral presentations. After meeting again in 1985 in Iowa City, the society chose to rotate annually between different venues. The original officers were the following: Edward Mason, MD, president; Boyd Terry, MD, secretary-treasurer; and Patrick O’Leary, MD, program committee chair. Initially, terms of office were 2 years in duration. However, as the work of running the society increased, the term of office decreased to 1 year in 1989 for the term of Cornelius Doherty. In recognition of his leadership in bariatric surgery, the Edward E. Mason Founders Lecture was established and given for the first time on June 2, 1989, by H. William Scott Jr., MD, from Vanderbilt. In the early days of the academic effort to define the science of the surgical treatment of obesity, there was support 49 by the NIH in convening a consensus conference and presentations of critical data at the Society for Surgery of the Alimentary Tract, the American Surgical Association, and the Western Surgical Association meetings. Many of the early surgeon scientists were also active in the American College of Surgeons (Ward Griffen, MD, and Patrick O’Leary, MD). The majority of surgeon leaders from this era commented on the serious barriers in trying to mainstream their research and surgical treatment of obesity into their departments and community hospitals. The formalization of the society served to promote the ability to provide a forum for exchange of ideas, research, and best practice and education of its members; however, it also established a political force within American surgery and served to promote access to care for the surgical treatment of obesity. The criticism and perception by the surgical establishment had a profound impact on the character of the society and drove some decisions (both good and bad) from a group that felt on the defensive. Perhaps, in many ways, this reflects the very real discrimination and prejudice that patients who suffer from obesity also feel. A fiercely independent and entrepreneurial character is firmly entrenched in the society’s foundation. These echoes of the underdog reappear throughout the 30+ year history of the ASMBS and continue to contribute to the development of our specialty and the identity of the society that serves to forward its practice. The nascent society continued to encounter a difficult environment full of opportunity. Cornelius Doherty, a private practice surgeon recruited to join Edward Mason in IU and president of ASBS from 1989 to 1990, writes: The early surgeons in our field operated at a time when the prejudice against surgical treatment of severe obesity was at its zenith. Organized medicine had abandoned them with indifference. Third-party payers were denying patient access to surgery arbitrarily. Professional liability carriers were stopping availability of coverage or charging exorbitant premiums. Plaintiff attorneys were predatory about filing cases. My agenda during my Presidency was to position the ASBS in the best possible way to plea the case for acceptance of surgical treatment of severe obesity at the National Consensus Development Conference of 1991. I had early notice that this conference would occur. I worked to that end tirelessly. I spearheaded the appointment of Lars Sjostrom as an Honorary Life Time Member of the ASBS. The team from ASBS effectively presented decisive data that advanced the recognition of the value of bariatric surgery. Twelve of the 14 surgeon speakers at the 1991 NIH Consensus Development Conference, “Gastrointestinal Surgery for Severe Obesity,” were ASBS members. In a breakfast meeting at Brennan’s in New Orleans, Michael Sarr, MD; Edward Mason, MD; John Kral, MD; Patrick O’Leary, MD; Cornelius Doherty, MD; and Harvey Sugerman, MD, set the agenda for the conference. These surgeons were able to present compelling data that influenced 50 R. P. Blackstone the panel of nonsurgical experts to express a positive overall Table 4.1 Presidents of ASBS/ASMBS position in the Consensus Conference Statement, which 1983–1985 Edward E. Mason, MD, PHD (A) John D. Halverson, MD (A) paved the way for improved acceptance of gastric restrictive 1985–1987 J. Patrick O’Leary, MD (A) or bypass procedures for patients affected by severe obesity 1987–1989 1989–1990 Cornelius Doherty, MD (PP/A) and influencing third-party payers. Thus, in many ways, 1990–1991 George S. M. Cowan Jr., MD (A) advocacy for access to care was involved in the original man1991–1992 John H Linner, MD (PP) date of the society. The society was focused during this time 1992–1993 Boyd E. Terry, MD (A) on getting at least 200 surgeon members so that they could 1993–1994 Otto L. Willibanks, MD (PP) qualify as a registered society with the American College of 1994–1995 Mervyn Deitel, MD (A) Surgeons. There was significant growth in the specialty of 1995–1996 Alex M. C. MacGregor, MD (PP) MBS, especially with the broader knowledge by patients that 1996–1997 Kenneth G. MacDonald (A) S. Ross Fox, MD (PP) there may be an effective treatment for this disease. The NIH 1997–1998 1998–1999 Henry Buchwald, MD, PhD (A) consensus conference was the pivotal event of this time and 1999–2000 Latham Flanagan, Jr. MD (PP) has stood the test of time. The 1991 guidelines still provide 2000–2001 Robert E. Brolin, MD (A) the backdrop against which decisions are made about 2001–2002 Kenneth B. Jones, MD (PP) whether a patient has access to surgery both in the United 2002–2003 Walter J. Pories, MD (A) States and around the world. 2003–2004 Alan C. Wittgrove, MD (PP) The society leadership reflects a strong commitment to 2004–2005 Harvey J. Sugerman, MD (A) both private practice and academic practice. Although there 2005–2006 Neil Hutcher, MD (PP) has never been a formal ratio established in the bylaws, tra- 2006–2007 Philip R. Schauer, MD (A) ditionally one-half of the Executive Council has come from 2007–2008 Kelvin Higa, MD (PP), first president of ASMBS Scott Shikora, MD (A) private practice with a rotation of the presidency from 1 year 2008–2009 John Baker, MD (PP) to the next between academic and private practice. As more 2009–2010 2010–2011 Bruce Wolfe, MD (A) surgeons in private practice have been publishing peer-­ a 2011–2012 Robin Blackstone, MD (PP) reviewed literature, serve to teach and train residents and fel2012–2013 Jaime Ponce, MD (PP) lows, participate in the quality program, and serve on and 2013–2014 Ninh T. Nguyen, MD (A) lead committees—along with the requirement by many aca2014–2015 John Morton, MD (A) demic surgical departments for high-volume practice and the 2015–2016 Raul Rosenthal, MD (A) employment by major hospital systems of physicians—the 2016–2017 Stacy Brethauer, MD (A) lines delineating private practice from academic practice 2017–2018 Samer Mattar, MD (A) have blurred. There remains, however, a very strong belief 2018–2019 Eric DeMaria, MD (A) that both aspects of practice should be represented in the A academic, PP private practice decisions of the society. Surgeons drove some of the pivotal aOnly woman to serve as president in the history of the society developments in the specialty in private practice (Table 4.1). of the data in bariatric surgery showed a lack of rigor; there was a growing public awareness of the increase in the numbers of patients with obesity as well as the number of surgerThe Era of Rapid Growth 1989–2004 ies being done for obesity. These challenges foreshadowed The original structure of the society was established in the the next era of the society’s growth. bylaws and has evolved throughout time. Officers were nomAt the time Boyd Terry, MD, became president (1991– inated by a nominating committee, and except for two elec- 1992), the society had just struggled through a major schism tions, the slate of officers was unanimously selected. The of its membership because of problems regarding the use of first occurred when Harvey Sugerman, MD, was nominated, dues for the journal and bylaw uncertainty. In addition, conbut George S. M. Cowan, MD, was elected in 1989. troversy between biliopancreatic diversion and duodenal This period of the society saw tremendous growth in the switch lent itself to spirited debate. Through Dr. Boyd’s leadnumbers of procedures and diversity of procedures including ership, the society emerged with more focus on representathe use of devices in large numbers of patients. Increasing tion from different regions of the country and an emphasis on numbers of surgeons operating without a knowledge base or communication and well-focused objectives for committee programmatic structure led to an increase in complications work. Surgeons within the society were concerned that their with a rise in malpractice premiums. Many insurance com- success with gastric bypass would be eroded by the adoption panies dropped benefits due to the sharp upturn in cost. This of untested “extreme” procedures that caused more harm than was demonstrated starkly in 2005 when the State of Florida good. This theme, existing in 1991, has had an echo throughlost all access for bariatric surgery by any company. Scrutiny out the history of the society. Intense procedural controversy 4 The History of the American Society for Metabolic and Bariatric Surgery erupted again when the Food and Drug Administration (FDA) approved the first device to be used in the treatment of obesity: the adjustable gastric band (AGB). With the increase in public scrutiny, surgeons who practiced unproven technology outside of Institutional Review Board (IRB) guidance came under increasing scrutiny and pressure not to offer unproven and untested procedure variations. The tendency to develop and use procedures without scientific support contributed to uncertainty by medical and surgical colleagues and patients. It hampers the advocacy by surgeons to garner support with payers who may believe that we are advocating surgery in order to line our own pockets. Even when we present valid and strong evidence, we have trouble convincing payers and others, in part because of this historical context. This tension between commercialism and scientifically based procedure indications continues to the modern era of the society including the omega-loop gastric bypass and one anastomosis duodenal switch. The society has taken a firm stand on these issues, discouraging the use of procedures that do not have sufficient evidence of safety and effectiveness from being performed outside IRB guidance. The society, led by datadriven analysis through the clinical issues committee and sanctioned by the Executive Council, developed and implemented a process for evaluating procedures and determining when the evidence is sufficient for the society to place them on the accepted procedures list. This encourages surgeons and industry who contribute to the creation of new procedures to work through an IRB process and establish an evidence basis prior to widespread implementation. ASBS began to achieve its political goals of formal participation in American surgery when it was voted a membership in the American College of Surgeons Board of Governors 1998. Patrick O’Leary, MD, had just joined the Executive Council of the Board of Governors, and when the request by Henry Buchwald, MD, came through, he was pivotal in getting it approved. Still, there were substantial barriers in the academic world. Particularly harsh was some of the criticism coming out of the surgical leadership of the University of Louisville, Kentucky, where one prominent surgeon declared bariatric surgery “charlatanism.” Within the academic establishment, surgeons who were involved in the surgical treatment of obesity were not well respected, their papers were not given credibility or even published, and their careers were at risk. Henry Buchwald, MD, recounts that when he became the president of ASBS, his chairman commented, “You have just killed your career.” Integrated Health Early on, awareness of the critical input and support of a variety of professionals in addition to surgeons were recognized. This was followed by the formation of the Allied 51 Health Sciences Committee (AHSC) in June 1990 with Georgeann Mallory, RD, as the first chair. The committee included registered dietitians, exercise physiologists, bariatric physicians, psychologists, nurse practitioners, and physician assistants. The membership of this committee, which elects its own president and council, has grown. Contributions both to the peer-reviewed literature and to clinical pathways of care as recognized in the accreditation standards have emerged to enhance the management of patients before, during, and after surgery. With the growing needs of the society, Georgeann Mallory, RD, who worked with Dr. Alex Macgregor, the 10th president of the ASBS, was appointed as executive director in 1993. She also served as the first chair of the AHSC. Mary Lou Walen was appointed the second chair of the AHSC. She writes: As Chair of the AHSC, it was important to me that all those working with patients receive education and information about the operations; complications; all aspects of care including working with the hospital both clinical and administration; learning about how to get paid for treating the patients; involving the primary care physicians and the specialists in becoming members of the treatment team; keeping the patients motivated and fully informed. During Walen’s chairmanship, workshops were developed and included in the program on clinical issues, patient education, insurance challenges, nutrition, psychology, and other topics; an allied health keynote speaker was added to the program; the AHSC chair was invited to all ASMBS Executive Council meetings; the committee requested to become a section and the Allied Health Sciences Committee became the Allied Health Sciences Section; and the president of the section became an elected position serving a 2-year term (Table 4.2). The AHSC chair became a voting member of the Executive Council of ASBS in 2004. The Allied Health Sciences Section became the Integrated Health Section in 2008. In the immediate perioperative period, the role of nursing in successful recovery and recognition of developing complications was recognized, and a formal test and certification in bariatric nursing for RNs working for two or more Table 4.2 Integrated health presidents of ASBS/ASMBS 1991–1996 1996–1999 1999–2004 2004–2006 2006–2008 2008–2010 2010–2012 2012–2014 2014–2016 2016–2019 Georgeann Mallory, RD Mary Lou Walen Tracy Martinez, BSN, RN, CBN Deborah Cox, RN Bobbie Lou Price, BSN, RN, CBN William Gourash, MSN, CRNP Laura Boyer, RN, CBN Karen Schultz, NP Christine Bauer, MSN, RN, CBN Karen Flanders, MSN, ARNP, CBN 52 years in the field was established through the leadership of Bill Gourash, PHD, MSN, CRNP, and a dedicated group of item writers. For this work Dr. Gourash received the inaugural ASMBS Integrated Health Distinguished Advanced Practice Provider Award. The Allied Health Section also established the Circle of Excellence Award, given annually to recognize outstanding ASMBS members who made contributions to the Integrated Health Section. The Integrated Health Section has been integral to incorporating the role of a multidisciplinary team into the requirements for accreditation in metabolic and bariatric surgery. The presidents of the Integrated Health and the integrated health council have played a significant role in developing an IH strategic plan. A focus over the last few years has been to update the nutrition guidelines and begin to share best practice by publishing tool kits for integrated health teams across the United States to use. They also have published a support group manual. They participate in every committee of ASMBS as well as having committees specifically for topics pertinent to the integrated health team members. Since the last publication of the history of ASBMS in the inaugural textbook, the Integrated Health Leadership has the following accomplishments: • Micronutrient guidelines • Support group manual • Webinar offerings for IH members (to provide education for all, but for those who cannot or do not attend OW) • Established an Integrated Health Facebook group and Twitter presence • A new Certified Bariatric Nurse web-based platform for renewals to simplify the process • CBN working toward accreditation as a certification program • >1000 Certified Bariatric Nurses • A task force exploring credentialing for advance practice providers • Tool kit on the ASMBS website with documents geared toward helping new (and experienced) providers with program start-up and development • YouTube videos on the value of integrated health individuals in ASMBS membership • Developed new categories of awards for recognition: Distinguished Behavioral Health Provider, Distinguished Advanced Practice Provider, IH Committee of the Year • Change in leadership (IH President, IH President-elect, and IH Secretary) terms from 2 to 1 year for each position, as well as change in process in which election is for IH Secretary who then rotates to IH President-elect who then rotates to IH President R. P. Blackstone Growth of the Society The rapid growth in the society paralleled the growth in the numbers of procedures and programs. This phenomenon was promoted by an increase in the number of people experiencing obesity, a growing awareness of surgical treatment of obesity, including the effect on type 2 diabetes; multiple stories began to be published including testimonials by celebrities like Carnie Wilson. The most significant factors in the growth of MBS were the transition from open to laparoscopic access with a resulting marked decline of mortality and morbidity and coverage by Medicare. Coupled with the implementation of national accreditation in the field and a strong access to care effort by the society, the numbers of people accessing surgery began to number in the tens of thousands. As the numbers of cases started to grow, the strongest focus of the society during this era was in the education of the membership. Dr. Brolin, who was president during the beginning of the “golden age” of laparoscopic access to bariatric procedures, focused on the training of general surgeons including a preceptorship committee (formed in 1999), which has evolved into the Bariatric Training Committee. The “golden age” of laparoscopic approach to bariatric surgery was born with controversy. By 2001–2002, during the presidency of Ken Jones, MD, the surgeons supporting open procedures and the surgeons who supported laparoscopic procedures were openly antagonistic to each other’s approach. Surgeons who had been doing open procedures were going to weekend courses sponsored by industry to learn the laparoscopic approach and coming back to their hospitals to do very complex laparoscopic gastric bypass procedures (GBP) with serious complications. At this time the delineation of privileges at many hospitals did not include advanced vs. basic privileges in laparoscopy. Since that time this has been corrected in part through the leadership of the Society of American Gastrointestinal and Endoscopic Surgeons (SAGES). This was a perfect storm for plaintiffs’ attorneys and the media, who regarded the surgery as unnecessary to treat obesity, a condition that they believed was self-inflicted by an inability to control one’s desire to eat. All of these aspects provided fertile ground for a malpractice crisis that almost brought down the society and the specialty. Once again, the society found itself on the defensive. This crisis was precipitated by a number of untrained general surgeons rushing into the then-fertile financial ground of providing bariatric procedures without appropriate training or structure. Led by Samar Mattar, MD, this evolved into a certification of fellows in MBS including a didactic study program and test. This program has provided a strong scientific and technical foundation for postgraduate practice of MBS. 4 The History of the American Society for Metabolic and Bariatric Surgery Medical Liability In the 1990s and into 2000, bariatric surgeons were making “news,” not so much for the benefits in health and quality of life for many but with not-so-back-page stories of procedures and outcomes gone awry for the few—especially for those cases or patients with notoriety. For some liability insurers, bariatric surgery outcomes were so uncertain that risk stratification of bariatric procedures resulted in regions where malpractice insurance for surgeons who practiced MBS was unavailable (Florida) or, if so, at premium rates that were increasingly higher than general surgery coverage. In 2005, with the support of our society, NOVUS Insurance Company, a risk retention group, was founded, with the expectation that with the guidance of a firm, expert in medical malpractice defense, bariatric surgery could be shown to be of actuarial risk similar almost to that of general surgery. It was clear to the Board, which was made up of regular members of the ASMBS that for any bariatric practice, careful attention to patient selection, education, evaluation, and operative preparation was critical. In addition, the consent process, with expanded face-to-face explanation and significantly improved documentation, was imperative. However, the basis for most lawsuits had to be recognized to have resulted not from technical operative error but a breakdown in patient, sometimes family, and physician rapport and untimely or inappropriate response to indicators of patient deterioration. In short, it is not that a leak occurred that makes a claim likely, and perhaps difficult to defend, but it is the “aggravating circumstances” together with an unanticipated outcome that make a claim virtually certain. Such circumstances are many, including surgeon unavailability when needed, inadequate surgeon empathy in a time of crisis, inexperienced “coverage” or poor “hand-off,” delay in or failure to respond to calls, lack of communication between all care providers, and inadequate initial risk disclosure paired with undocumented patient understanding. In addition, there is a well-established and documented weight bias among health-care providers and within the health-care industry toward not only the patient with obesity but also the surgeons or physicians who treat it. The society realized that a strategy to establish a quality standard as well as share best practice would be necessary and would be necessary to underlie the overall increase in case volume. In 2011, NOVUS was merged into NORCAL Mutual Insurance Company. In 2011, the ASMBS “Professional Liability” Committee became the “Patient Safety” Committee. Increasingly, the committee recognized the importance of closed claims as a significant resource in our improving patient safety. In 2012, our monthly e-publication (“Top 5 on the 5th”) vignettes, derived from closed claims, met with broad society support. They are anticipated to 53 resume in a new format in the ASMBS new magazine Connect. Investigation is ongoing as to whether we may develop a closed claims database, similar to that of the American Society of Anesthesiologists, which for more than 30+ years has resulted in material improvements in anesthesia services and a 30% average decline in anesthesiologists’ liability premiums. Over the past 5 years, more work has done on evaluating closed claims, which were collected in 2016 and 2017. he Era of Quality and Engagement T from 2004 to Present Quality and Data Registries One of the outgrowths of the period of crisis from 2001 to 2004 was an awareness that the image of the society needed to change. Rather than allowing any surgeon with minimal training, low volume, or no programmatic elements to participate, the society made a decision in its annual business meeting to establish a national center of excellence program. A minimum of 125 cases was required to qualify with the result that the number of operating surgeons and programs contracted sharply throughout the next few years. Surgeons and programs that did not participate lost their ability to offer MBS as the contraction in the market place occurred. As part of that effort, the Bariatric Outcomes Longitudinal Database (BOLD) was developed to support the accumulation of data for both outcomes’ information and research. This was not, however, the society’s first efforts at creating a registry. Standardized data collection and analyses for surgical treatment for obesity began in 1985 under the direction of Edward Mason, Department of Surgery, University of Iowa Hospitals and Clinics (UIHC). The National Bariatric Surgery Registry’s (NBSR) goal was to meet a growing need for quality control in reporting outcome results by assisting surgeons in continuing improvement for patient care through outcome analyses. The NBSR was run in the Department of Surgery at UI and received full financial support from one corporate sponsor during the first 2 years. Subsequent support came from surgeons who voluntarily participated through membership fees, satellite data collection, and submission. The International Bariatric Surgery Registry (IBSR) provided software, training, and instruction manuals for collecting, storing, and preparing reports of local data for comparison with the total data reported. Management of the system was by Kathleen Rehnquist, BS, from 1986 to 2006. Dwight “Ike” Barnes; John Raab, RN; and Mark Crooks provided personal computer programming. Together they provided successful software throughout ten updates/revisions. Graduate students provided the integrated statistical analysis from the College 54 of Preventive Medicine, biostatistics division (Donald Jiang; Elizabeth Ludington, PhD; Wei Zhang, PhD; and Shunghui Tang, PhD). Professors mentoring the students included Robert Woolson, PhD; Miriam B. Zimmerman, PhD; and Michael P. Jones, PhD. The aggregate analyses were accomplished via SAS programming at the University Computer Center on IBM mainframes until aggregate analysis and computers evolved to use PC SAS on a personal computer housed in the IBSR office. In 2006, the final repository of the aggregate data represented 85 data collection sites for 45,294 subjects whose surgery was performed by 148 surgeons many of whom were members of ASBS. Newsletters, manuals, papers, and data for lectures or publication could be prepared using the IBSR software or by special reports of the aggregate data with assistance from IBSR staff. Direct access to the registry data was never available, due to privacy policies of the UIHC, State of Iowa, and Federal Regulations (HIPPA). Quarterly reports were provided to each satellite surgical practices with de-identified results to help surgeons compare patient outcome with that of the total IBSR experience. More than 70 newsletters were published, with Dr. Mason soliciting a medical section for surgeons and other IBSR staff writing articles of interest regarding data collection and how data results were reported. The ultimate closure of the IBSR resulted from inadequate financing to support a Web-based data collection system and incomplete follow-up methods for complete data analysis and verification. BOLD was the society’s second effort at a registry. In 2004 the ASBS established an independent not-for-profit company, the Surgical Review Corporation (SRC), to oversee the ASMBS Centers of Excellence quality program. The BOLD registry was developed with input by ASMBS surgeons. A few years after the COE program was implemented, participation in the registry became a requirement for accreditation. Requiring data entry of all bariatric cases began the process of changing the surgical culture within community hospitals where the majority of patients received MBS. This second effort at a data registry was also problematic. These include data not available reliably for use in continuous quality improvement; individual surgeon office, rather than hospitals, often paid registry fees and collected; poor definitions and haphazard methods; and quality of data collection and poor long-term follow-up. In addition, the registry was not connected to the national data quality movement and began to fall behind other efforts like the National Surgical Quality Improvement Program (NSQIP). Despite the important efforts of Deborah Winegar, PhD, the final database director and surgeon members of the quality program who worked tirelessly to try and improve the registry, a point was reached which required a change in direction. A scientific project to compare three data registry options was conducted, and with input from the ASMBS Quality and Standards Committee, R. P. Blackstone the society decided to move away from BOLD. The aggregate data from BOLD has not been lost; some publications have resulted from this data. Under the leadership of Robin Blackstone, MD, President of the ASMBS, and David Hoyt, Executive Director of ACS, ASMBS joined the Centers of Excellence program with the American College of Surgeons Bariatric Surgery Network on April 1, 2012. This established one authority for national accreditation in the United States, the Metabolic and Bariatric Surgery Accreditation and Quality Improvement Program (MBSAQIP). The program established a registry based on strong principles of data collection and integrity of the data, including collection of 100% of cases performed at the accredited center; defined data entry variables; and thirdparty data collection by certified of the clinical reviewers. Each program receives two semiannual reports (SAR) that allow comparison of their programs on key variables to national benchmarking providing the high-quality data necessary for the use of outcomes to improve practice at the local level with continuous quality improvement. In addition to local efforts, the data is examined by the MBSAQIP Quality Committee to identify areas where national improvement projects could take place. One project focused on programs with high emergency department readmission rates. In this project, led by John Morton, MD, programs that were high outliers in readmissions were identified and offered an opportunity to participate in the DROP process (Decreasing Readmissions through Opportunities Provided) [2]. The second project, led by Stacy Brethauer, MD, offered programs with higher length of stay the opportunity to participate in the use of a set of enhanced recovery protocols for metabolic and bariatric surgery. In addition to prospective projects that occur in the real-world setting of community practice as demonstrated by these two national quality improvement projects, the MBSAQIP registry accumulates high numbers of patients annually. The quality of data collected and the availability of public use files of the previous year’s data allow retrospective review and study of even small effect size complications. The data registry provides high-quality data that offers a credible foundation for quality improvement and best practice at the community and university hospital level. Strategic Plan Development The society has successfully met many challenges during its history. However, the world of medicine changes constantly, and in order to respond, the society created a plan for its own evolution. A formal strategic plan for the society—led by Phil Schauer, MD, with input from the Executive Council— was developed and embraced at the business meeting in 2008. As part of the evolution of the specialty, the society 4 The History of the American Society for Metabolic and Bariatric Surgery elected to change its name from the American Society for Bariatric Surgery (ASBS) to the American Society for Metabolic and Bariatric Surgery (ASMBS) at the annual business meeting on June 15, 2007. The strategic plan was implemented fully during the presidency of Bruce Wolfe, MD, with a change in the structure of the committees to align with the established mission, vision, values, and goals of the society and direct alignment of the committee projects with the overall budget of the society. The strategic plan drove many of the expenditures of the society, and all budget items are considered in light of the overall mission and goals. The alignment of the committee structure enabled a much higher productivity in the committees and drove improved communications and work product of the committees. Every facet of the society from budget decisions to the overall work plan of the committees was aligned. Dr. Wolfe also created a Quality and Standards Committee to assess the accreditation program and propose an evolutionary process. These updates to the operating structure of the ASMBS would transform the society into one that had the engagement of a very large group of young leaders and members of the society from both academic and private practice. This current model has provided robust volunteerism and energized committees with emerging and diverse merit-based leadership. Implementing a culture of leadership development has been the ultimate guarantee of continuation of new ideas and strategy to meet future challenges. The current committees report evolving goals/objective and accomplishments each year in the ASMBS Annual Report (https://asmbs.org/about/annual-report). The Journal Obesity Surgery, the original journal of the ASBS, was founded in 1990, adopted by the society in 1991 and achieved Index Medicus status in 1995. Its birth was not without controversy. In 1989 at the annual meeting in Nashville, Tennessee, the Executive Council, at its statutory meeting, had prepared a nominating slate for consideration at the business meeting, including a proposal initiated by the president, Patrick O’Leary, MD, for joining the North American Association for the Study of Obesity (NAASO) and other International Association for the Study of Obesity (IASO) organizations in adopting the International Journal of Obesity as the ASBS journal. As a guest at the council meeting, Dr. Mervyn Deitel presented his own proposal for a journal. The Executive Council supported Dr. Deitel’s proposal, pending his providing a business plan and other details. At the business meeting, however, following controversial presentations during the scientific meeting, which drew criticism for premature clinical use of novel operations without adequate patient follow-up, the membership rejected the 55 nominating committee’s slate of candidates. At that same meeting, the membership voted on, and accepted, the proposal to adopt IJO as its journal, affirming the possibility that, at some later time, Dr. Deitel might provide a separate proposal to be duly considered. A few months later, Dr. Deitel (with support from some newly elected council members) mailed selected members requesting support for his journal. His rallying cry: “Pull out all your rejected manuscripts and we will publish them!” Through a closed ballot process, the leadership of ASBS nullified the 1989 business meetings’ decision, adopting Dr. Deitel’s journal with mandatory subscription in 1990. Dr. Deitel, who had sole ownership of the journal, later decided to sell the journal. It came as a surprise to younger members that the journal Obesity Surgery was actually privately owned; after considering the option to purchase it, the ASBS decided to establish its own journal. Eventually, Obesity Surgery was sold to a publisher and adopted as the official journal of the International Federation for the Surgery of Obesity and Metabolic Disorders (IFSO). It maintains the tradition of publishing articles from the international community and—under the direction of Dr. Henry Buchwald and Nicolas Scopinaro, MD, and the current editor, Scott Shikora, MD—has made significant progress in improving its impact factor. In February 2004, the ASMBS established a new journal Surgery for Obesity and Related Disease (SOARD) owned by the society—with Harvey Sugerman, MD, as the first and current editor. Dr. Sugerman is largely credited with developing an outstanding editorial board and with the high quality that the journal has achieved. The initial journal was published in six issues during the year, increasing to monthly publication in 2017. During the course of the journals’ history, the editorial board has made concerted efforts to standardize the reporting of key variable like total weight loss, in order to produce articles with less heterogeneity. The impact factor in 2017 was 4.5, placing it as number 11/165 surgical journals. In 2017, the journal published 396 original manuscripts. The journal represents the readership with 44% of manuscripts from North America, 34% from Europe, and 10% from Asia. A continuing medical education program was implemented and led by Samar Mattar, MD, awarding 3671 h of CME to 278 readers and 472 h of CME to reviewers in 2017. Dr. Sugerman continues to lead the journal effort. Raul Rosenthal, MD, was appointed coeditor. The 14-year history of the journal is a tribute to the men and women who design, execute, and write about their research and to those who guard the portals of good science in ensuring the journal reflects the highest values of inquiry. The journal allows us to bring the light of scientific inquiry to shine on our work in an environment without the bias that Dr. Sugerman and many others, who struggled for recognition of 56 their work during their careers, faced. The journal is the embodiment of how far the specialty has traveled. Access to Care Surgeons who treat other forms of disease have enjoyed wide access to their procedures through coverage by insurance. Patients who suffer from obesity, however, have long been victims of a misperception of their role in being affected by obesity (personal responsibility) and denied coverage based on the perception of the “cosmetic” nature of surgical treatment. This quest to obtain wider access for patients has been one of the critical driving forces behind the society’s growth. These efforts have been ongoing since the earliest days of the society but were formalized by the creation of the Access to Care Committee on November 11, 2008. During the next 5 years, the work of the committee included partnering with advocates outside ASMBS in the battle for access. This coincided with the vision the society had to create a population management approach to the management and treatment of obesity. Part of this strategy is to provide more balance in reporting around obesity and to train advocates at many levels of leadership with ASMBS. The society engaged Roger Kissin and Communications Partners in order to fulfill the strategic goal of making the society the public voice of authority in this field and add to the education of the media about this subject. This strategy has been extremely successful in changing the dialogue. Currently, the president and senior leadership give more than 300 interviews to major media outlets per year with messaging that is developed and approved by the Executive Council; media training is provided to all committee chairs and chapter presidents so that when we respond to a query, we can do that with one consistent message. Another successful strategy was to field a rapid response team approach to changes in benefits. If any entity (company, government agency, state agency) tried to change or drop a benefit or began to consider implementing one, a group of experts—including the surgeons from that area, industry with lobbyists on the ground, and leadership from the ASMBS Access to Care Committee as well as the Obesity Action Coalition (OAC)—could convene to immediately address the problem. This has been a very successful strategy in maintaining and gaining new coverage. The most convincing argument, however, is the effectiveness of surgical therapy both on obesity itself and, perhaps even more profoundly, for the effect on obesity-related diseases such as diabetes. Even with all these efforts, which are intense and ongoing, far less than 1% of the patients who have significant disease that will limit their longevity have access to the most effective care. Although we often think of access as limited by R. P. Blackstone coverage, in fact it is just as limited by the available surgical manpower, which at this time can provide only 1% of patients with surgical treatment. It is also limited by reimbursement. It takes many resources in structure, process, and personnel to support patients through the entire course of care, and reimbursement for all of this supportive care is lacking. In this environment, the tricky questions of who should have access to surgery and what the optimal procedure should be persist despite efforts to define indications. Meanwhile the scientific data on epigenetic transfer of obesity-promoting genes and the differences in physiology in regard to hunger, satiety, and metabolism of patients who suffer from obesity are now widely documented. Support by the government for treatment came with the announcement of the National Coverage Decision by the Centers for Medicare and Medicaid Services (CMS) providing access to surgical treatment for Medicare and Medicaid beneficiaries as of February 21, 2006. It was expanded for the treatment of diabetes in 2009. Although CMS continues to support access to surgery, they dropped the requirement for accreditation of the program providing the surgical care—a decision born in controversy and of great concern to the society about the safety of the decision. Currently the fight for access continues and has come down to a state-by-state battle to establish bariatric surgery as an essential benefit in the Affordable Care Act. Twenty-two states recognize bariatric surgery as an essential health benefit. All these politics are local, and a local political force is needed. To meet this need, the ASMBS state chapter program was established during the presidency of Neil Hutcher, MD. The goals for the state chapters are to advocate for increased access at the local level and to establish collaboration for best practice in the quality program. Each state chapter has an elected State Advocacy Representative. At the beginning of 2018, OMA and TOS announced that their respective groups would be establishing State Advocacy Representative (STAR) Programs—modeled after the ASMBS STAR Program. Both TOS and OMA are hopeful that they will have a STAR in every state by the end of 2019. OAC is also formulating plans for regional OACSTARs. At the time of this report, plans were underway to establish an Obesity Care Continuum STAR Program to link these programs across AND, TOS, OMA, OAC, and ASMBS. Obesity Action Coalition In addition, a need for advocacy on the policy level was identified. Experts on public policy from Kellogg School of Business at Northwestern were engaged to study the access problem. Led by Dr. Daniel Diermeier, it was determined the nature of the issues that led to the loss of insurance coverage required a public policy approach. The Obesity Action 4 The History of the American Society for Metabolic and Bariatric Surgery Coalition (OAC) was founded in 2005 by Robin Blackstone, MD; Georgeann Mallory, RD; and Christopher D. Still, DO, FACN, FACP, to fill the patient-advocacy gap for the disease of obesity. The Obesity Action Coalition (OAC) is a more than 60,000 member-strong 501(c) (3) national nonprofit organization dedicated to giving a voice to the individual affected by the disease of obesity and helping individuals along their journey toward better health through education, advocacy, and support. The OAC’s core focuses are to raise awareness and improve access to the prevention and treatment of obesity, provide science-based education on obesity and its treatments, fight to eliminate weight bias and discrimination, elevate the conversation of weight and its impact on health, and offer a community of support for the individual affected. The OAC is made up of a vibrant membership community where individuals can find valuable information to help them on their weight journey and connect with others who share similar experiences. The OAC is also the founder of the highly successful Your Weight Matters brand, which encompasses Weight Matters Magazine, the Your Weight Matters National Convention, and the Your Weight Matters National Campaign. The goal of the Your Weight Matters brand is to deliver one clear, concise message: “Your Weight Matters – For Your Health.” Obesity Care Advocacy Network The leading obesity advocate groups founded the Obesity Care Continuum or “OCC” in 2010 to better influence the health-care reform debate and its impact on those affected by overweight and obesity. The OCC was composed of the Obesity Action Coalition (OAC), the Obesity Society (TOS), the Academy of Nutrition and Dietetics (AND), the American Society for Metabolic and Bariatric Surgery (ASMBS), and the American Society for Bariatric Physicians (ASBP). The American College of Surgeons, although not a member of the OCC, supports the work of the group by acting as an independent third-party advocate. The OCC evolved into the Obesity Care Advocacy Network (OCAN) with a mission to partner with medical societies and organizations to change how the nation perceives and approaches the US obesity epidemic by educating and advocating for public policies and increased funding for obesity education, research, treatment, and care. Now with a membership of 19 organizations including the American Academy of Physician Assistants; Academy of Nutrition and Dietetics; American Association of Clinical Endocrinologists; American Association of Nurse Practitioners; American Council on Exercise; American Society for Metabolic and Bariatric Surgery; AMGA; Black Women’s Health Imperative; 57 Healthcare Leadership Council; National Alliance for Healthcare Purchaser Coalitions; Novo Nordisk, Inc.; Obesity Action Coalition; Obesity Medicine Association; SECA; the American Gastroenterological Association; the Endocrine Society; The Obesity Society; Weight Watchers; and the YMCA of the United States. The group has sponsored three workgroups centered around implementing the provisions of TROA through the administrative mechanisms as well as address military readiness on the impact of obesity on the national armed forces. The Treat and Reduce Obesity Act (TROA) In June of 2013, the ASBMS, as part of the OCC network, endorsed the Treat and Reduce Obesity Act of 2013 (TROA)—a bipartisan, bicameral bill that has been introduced in the 113th Congress. The bill aims to effectively treat and reduce obesity in older Americans by increasing Medicare beneficiaries’ access to qualified practitioners who can deliver intensive behavioral therapy for obesity and allowing Medicare Part D to cover FDA-approved obesity drugs. The initial efforts to enact the bill were not successful; however, it was reintroduced into congress in April of 2017 by Senators Bill Cassidy (R-LA) and Tom Carper (D-DE) and Representatives Erik Paulsen (R-MN) and Ron Kind (D-WI). TROA (Senate Bill 830/House of Representatives Bill 1953) is strongly supported by OCAN as well as the American College of Surgeons who joined the advocacy effort in 2018. Specifically, TROA will provide the Centers for Medicare and Medicaid Services (CMS) with the authority to expand the Medicare benefit for intensive behavioral counseling by allowing additional types of health-care providers to offer these services. The legislation would also allow CMS to expand Medicare Part D to provide coverage of FDA-approved prescription drugs for chronic weight management. The budget impact analysis paper developed by Wayne Su and IHS Markit has been useful in demonstrating the significant potential savings to the Medicare program ($19–21 billion) over 10 years should Congress pass TROA. ational Obesity Care Week (NOCW) 2018 N (October 7–13) The Obesity Action Coalition (OAC), The Obesity Society (TOS), the STOP Obesity Alliance, the Obesity Medicine Association (OMA), and the American Society for Metabolic and Bariatric Surgery (ASMBS) launched the first NOCW in 2018. The goal is to build within the public understanding of obesity and value of science-based care. The ASMBS 58 R. P. Blackstone believes that NOCW elevate awareness of the disease of obesity, create an understanding of the challenges people affected by obesity endure, and promote the support of treatment. International Affiliations Obesity is an epidemic affecting many countries outside the United States. Our colleagues from around the world have made exceptional contributions to the science and art of MBS. In recognition of this, ASMBS became a founding member of the International Federation for the Surgery of Obesity (IFSO) formed in 1995 at a meeting in Stockholm. IFSO currently has more than 50 member societies. There are 3600 members of ASMBS that are also members of IFSO. The international committee was organized in 2009, and Raul Rosenthal, MD, was the first chair. The first International Congress was at the 2011 Annual Meeting: Bariatric Surgery in Latin America. International interest and growth have been strong and increasing annually (Fig. 4.2). The ASMBS Foundation The ASMBS Foundation is a 501(c) (3) nonprofit organization developed to raise funds for conducting research and education, increasing public and scientific awareness and understanding, and improving access to quality care and treatment of obesity and severe obesity. The ASMBS Foundation was established through the efforts of the ASMBS Executive Council in 1997 spearheaded by S. Ross Fox, MD. The ASMBS Executive Council and Dr. Fox recognized the need to provide fund-raising— through charitable gifts and public and private donations—to support their shared vision to improve public health and well-being by lessening the burden of the disease of obesity and related diseases throughout the world. The foundation has continued to support the activities of the society centered around access, education, and research. Currently the foundation is undergoing a strategic process to revitalize the board, building a stronger foundation of philanthropy with a new executive director, and continuing to build national presence to the Walk from Obesity. Obesity Week In fall 2013, the ASMBS and the Obesity Society (TOS) held their annual meetings in conjunction with one another. Surgeons, researchers, bariatric medicine specialists, and ASMBS Historical Membership 2018 4500 4117 4065 4070 4000 4119 4247 3952 3800 3550 3500 3385 3059 3000 3198 2878 2697 2500 2407 2000 1500 1000 2385 1922 1511 1364 956 931 1619 1251 1078 1710 1833 1168 1226 1916 2055 2109 1282 1330 2576 2641 2180 1620 1441 2417 2462 2433 1732 1648 1608 1519 1543 1606 896 686 693 671 533 527 415 420 433 312 352 341 MD IH 263 177 192 188 244 264 274 304 159 163 168 102 218 233 TOTAL 48 74 113 26 26 31 38 0 14 24 1992 1993 1994 1995 1996 1997 1998 1999 2000 2001 2002 2003 2004 2005 2006 2007 2008 2009 2010 2011 2012 2013 2014 2015 2016 2017 2018 500 Fig. 4.2 Growth of the ASMBS 4 The History of the American Society for Metabolic and Bariatric Surgery integrated health professionals came together for one action-­ packed week. Each society maintained its own traditions and meetings, but each member who attended was able to choose from among a wide variety of educational options. Phil Schauer, MD (ASMBS), and Gary Foster, MD (TOS), forged the path to the first conference supported by the ASMBS Executive Council and membership. The meeting is designed to foster the understanding of the pathophysiology of obesity, the application of science to the clinical management of patents, and the mechanism of action of surgery and pharmaceutical and behavioral management and to establish collaboration in research. Obesity week has blossomed into one of the most well-attended scientific meetings of the year with an expanded catalogue of course offerings and sharing of dialogue among its many diverse members. Attendance in 2018 included 5300 people, equally divided between surgery and medicine physicians and integrated health members. Conclusion The history of the ASMBS is one of the intense and focused efforts by visionary leaders, but it is also the story of engagement of the members in the development of the specialty. The foundation of the society is grounded in the efforts of critical thinkers, scientists, and visionaries, but with the transition to the national accreditation system, all members of the society have participated in one of the most important and successful quality initiatives in American surgery. The sense of having a special mission, of championing a group of patients who face daily discrimination and prejudice, and of being fierce advocates for a science that has delivered hope to millions of patients affected by diabetes and obesity defines members of the ASMBS. The surgeons and integrated health colleagues of ASMBS deliver on a daily basis the most effective therapy in the history of medicine, with a mortality that is less than a laparoscopic cholecystectomy. The American Society for Metabolic and Bariatric Surgery has matured throughout the 30+ years of its existence. The society has shown visionary leadership in educa- 59 tion, multidisciplinary care, access to care, accreditation, and quality improvement. The ASMBS has responded to the crises of its time with action and become part of the wider society of physicians managing obesity. The society has taken a leadership position in defining approved procedures, providing guidance to the FDA in the approval of new devices, providing guidance to members on a wide range of topics, and establishing an ethics committee to hear grievances about advertising and practice issues. The twin drivers of access to care and quality have driven engagement of the membership with their society. The strength of the society lies in the adherence to scientifically valid principles, fairness, and increasing transparency of governance and in the engagement of talented members who volunteer their time to serve on committees. The dedication of our members to provide high-quality safe care continues to be our most closely held goal. Although we may have been considered outsiders at one time, our experience in quality and collaboration, access to care issues, and managing change should propel us into the leadership of our hospitals and American surgery. Acknowledgments Written in collaboration with the following group of contributors: Ed Mason, MD, FACS; Cornelius Doherty, MD; Patrick O’Leary, MD; George S. M. Cowan Jr., MD; Boyd E. Terry, MD; Henry Buchwald Jr., MD; Robert E. Brolin, MD; Kenneth B. Jones, MD; Walter Pories, MD; Harvey Sugerman, MD; Neil Hutcher, MD; Kelvin Higa, MD; Scott A. Shikora, MD; Karen Schultz; Laura Boyer, RN; Mary Lou Walen; John Kral, MD, PhD; Mal Fobi, MD; William Sweet, MD; George Blackburn, MD; James Zervios, OAC; Connie Stillwell, ASMBS Foundation; Georgeann Mallory, RD; Joe Nadglowski; Chris Gallagher; Kathleen E. Renquist; and Karen Flanders. References 1. Blackburn GL, The Edward E. Mason founders lecture: interdisciplinary teams in the development of “best practice” obesity surgery. Surg Obes Relat Dis. 2008;4:679–84. 2. Morton J. The first metabolic and bariatric surgery accreditation quality improvement program quality national initiative: Decreasing readmissions through opportunities provided. Surg Obes Relat Dis. (SOARD May–June. 2014;10(3):377–8. 5 Physiological Mechanisms of Bariatric Procedures David Romero Funes, Emanuele Lo Menzo, Samuel Szomstein, and Raul J. Rosenthal Chapter Objectives 1. Describe some of the most commonly accepted theories regarding the physiological mechanism of bariatric procedures. 2. Address the potential physiological mechanisms affecting both weight loss and resolution of diabetes. Introduction In spite of the fact that obesity has reached pandemic proportions, it remains one of the most neglected public health problems in the United States. This has awakened a growing interest in placing obesity as a prime subject of many recent public health campaigns. The need for these considerable efforts derives from the alarming reports of the prevalence of obesity in the US population. In 2015–2016, the prevalence of obesity was 39.8% in adults and 18.5% in adolescents [1]. Despite the increased awareness of its severity, the linear time trend forecasts suggest that by 2030, 51% of the US population will be obese [2]. The progressive and continuous rise in the prevalence of obesity have also determined a secondary epidemic of the related comorbidities, in particular the risks of cancer, diabetes, and cardiovascular diseases. Bariatric surgery is now considered the first management option for failure of medical treatment in severely obese subjects and the most effective method for sustained long-term weight loss. There is extensive evidence of the positive metabolic impact, remission, and resolution of most of the comorbidities associated with severe obesity, diabetes included [3]. Historically, bariatric procedures are thought to induce D. R. Funes (*) · E. Lo Menzo · S. Szomstein · R. J. Rosenthal Department of General Surgery, The Bariatric and Metabolic Institute, Cleveland Clinic Florida, Weston, FL, USA e-mail: funesd@ccf.org weight loss by causing caloric restriction and/or malabsorption. However, newer mechanistic studies, in parallel with the establishment of the gastrointestinal tract as a key regulator of energy and glucose homeostasis, have introduced the hypothesis that alternative mechanisms mediate the weight-­ reducing and metabolic benefits of most bariatric/metabolic operations. Furthermore, as the close interaction between diet, gut, and brain hormones unwinds, the mechanisms of action of these procedures, as well as their classification, have significantly changed. In fact, the pathway describing how the centrally regulated body weight homeostasis is profoundly influenced by hormones secreted in the intestinal tract and adipose tissue is now well recognized [4]. The overall balance of these peripherally secreted hormones and their interaction at the level of the hypothalamus would eventually affect food intake and energy expenditure [5]. The mechanism of diabetes resolution after bariatric surgery is not entirely understood. Since insulin resistance is one of the main etiologies, it seems obvious that weight loss, although not the only component, continues to be an essential contributor. In fact, typically diabetes improvement or resolution occurs within weeks after bariatric procedures. Regardless if it is gastric bypass (GBP), sleeve gastrectomy (SG), or biliopancreatic diversion (BPD), in all of these procedures, remission ensues from the preceding and expected weight loss [6, 7]. Pories et al. were the first to suggest that caloric restriction played a key role in the resolution of diabetes; following this historical finding, the important gluco-­ regulatory roles of the gastrointestinal (GI) tract were firmly established. However, the physiological and molecular mechanisms underlying the beneficial glycemic effects of bariatric surgery remain incompletely understood. In addition to the mechanisms proposed by Pories et al., currently other hypotheses involving changes in bile acid metabolism; GI tract nutrient sensing and glucose utilization, incretins, and possible anti-incretin(s); and the intestinal microbiome have gained strength. According to recent studies, these changes, acting through peripheral and/or central pathways, © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_5 61 62 D. R. Funes et al. lead to reduced hepatic glucose production, increased tissue glucose uptake, improved insulin sensitivity, and enhanced ß(beta)-cell function. These findings suggest that a constellation of factors, rather than a single domineering mechanism, likely mediates postoperative glycemic improvement, with the contributing factors varying according to the surgical procedure [8]. Nevertheless, all coincide in one common positive effect resulting in the resolution of diabetes. Here, we describe some of the most commonly accepted theories regarding the mechanism of action of the most widely accepted bariatric procedures. Mechanism of Action Caloric Restriction The current understanding of different mechanisms of action of these procedures, in particular the role of gut hormones, has led to dispute the traditional classification of bariatric procedures in the three main categories: restrictive, malabsorptive, and combined. Although a clear understanding of all the mechanisms of action of bariatric procedures has not been reached, multiple theories exist. It is likely that several factors contribute to the final efficacy of the procedures. Because of the overlap of effects, we will address the potential mechanisms of action affecting both weight loss and diabetes resolution. Potential contributors to weight loss and diabetes resolution are outlined in Table 5.1. Malabsorption As previously mentioned, the surgically induced alterations of the normal gastrointestinal absorption process lead to various degrees of weight loss. This is especially true in procedures such as the BPD and the BPD with duodenal switch (BPD-DS) where long alimentary (250–300 cm) and biliopancreatic limbs leave a short (100 cm) common channel for Table 5.1 Potential mechanisms of action of bariatric operations Mechanism of action Malabsorption Caloric restriction Energy expenditure ∆(Delta)-eating behavior Hormonal Vagus nerve Bile salts Adipose tissue Microbiota ß(Beta)-cell function Insulin sensitivity Procedure RYGB LSG ± − + + ± − + ± LAGB − ± − − BPD + + + ? BPD-DS + + + ? + ?/− + + ± ± ± − ?/− ± − − − +a + ?/− + + ± ± ++ + ?/− + + ± ± ++ Only related to weight loss a the absorption of nutrients. Even the more conservative alimentary limb lengths (100–150 cm) of the standard gastric bypass have been shown to create a certain degree of fat malabsorption, as demonstrated by the increase in fecal fat at 6 months (126%) and 12 months (87%) [9]. Since there is no significant alteration of the protein and carbohydrate absorption, the overall reduction of the combustible energy absorption has been shown to be only 6–11% [10]. While it is true that the more malabsorptive procedures (BPD, BPD-DS) result in a more impressive weight loss (excess weight loss [EWL], 79%) and diabetes resolution (98.9%), it is unlikely that the malabsorption by itself is solely responsible [11]. + ?/− + +a ? ? + Caloric restriction is one of the immediate effects of RYGB and SG due to anatomical changes. The beneficial effect of caloric restriction on the glycemic control has been previously demonstrated [12]. The carbohydrate-controlled calorie-­restricted diet produces up to 40% improvement of the insulin resistance and ß(beta)-cell function as measured by the homeostatic model assessment (HOMA) method in just 2 days [13]. If continued over a period of 11 weeks, the diet can improve the peripheral insulin resistance, even if the hepatic insulin sensitivity remains unchanged [13]. In the perioperative period of bariatric surgeries, the caloric intake is dramatically reduced to 200–300 kcal/day. This factor undoubtedly contributes to the immediate weight loss experienced by these patients postoperatively. In fact, some authors were able to demonstrate similar weight loss results in non-operated obese subject after 4 days of post-Roux-en­Y gastric bypass (RYGB) diets [14]. The rate of secretion of gastrointestinal hormones, however, was altered in the RYGB group [14]. These findings were replicated by other authors who found similar results in weight loss at short-­ term follow-up comparing RYGB and low-calorie diet, but only RYGB patients determined improvements of insulin resistance, insulin secretion, and insulin-stimulating gut hormones, such as GLP-1 [15]. This is obviously true only for the first few weeks. In fact, there is a significant difference in the rate of weight loss, as demonstrated by the time needed to lose 10 kg between RYGB (30 days) and caloric restriction (55 days) [16]. Also, if the caloric restriction was the only responsible mechanism for glucose control, the improvement of this parameter should be uniform between the different bariatric operations. It has been clearly demonstrated how BPD ± DS, RYGB, and laparoscopic sleeve gastrectomy (LSG) provide a quicker improvement of diabetes as compared to laparoscopic adjustable gastric banding (LAGB) [11, 17]. This is also demonstrated by the change in the profiles of the glucose and insulin curves between LAGB, low-calorie diets, and RYGB. In fact, if LAGB and a low-­ 5 Physiological Mechanisms of Bariatric Procedures calorie diet produce a downward shift of such curves, RYGB determines shortened times to peak glucose and insulin with a leftward shift of the curves [18]. It is reasonable to conclude that, although caloric restriction is an important factor contributing to the improvement in hepatic insulin sensitivity, it likely plays a role only in the immediate postoperative period, and other factors are involved in the long-term weight loss and glycemic control improvement. Energy Expenditure Under normal circumstances, the energy expenditure decreases consequently to caloric restriction and the resulting weight loss [19]. This adaptive mechanism on one hand is meant to preserve the individual and on the other hand could be responsible in part for the long-term failure of the caloric restrictive diets. The data on energy expenditure after bariatric surgery is somewhat conflicting. In fact, if some investigators found a decrease in energy expenditure secondary to the weight loss after RYGB, others were able to demonstrate its increase in both RYGB and BPD, but not after vertical banded gastroplasty (VBG) [15, 20–22]. There is data that shows evidence of significant reduction of resting energy expenditure and a significant degree of metabolic adaptation both occur after sleeve gastrectomy. The hypothesis is that a greater metabolic adaptation could be partly responsible for a lower weight loss after surgery. Furthermore, there is recent evidence that suppression in resting energy expenditure after sleeve gastrectomy and RYGB remained up to 2 years, even after weight loss had plateaued. This study suggests that energy adaptation is not a contributing mechanism to medium-term weight maintenance after SG and RYGB bariatric surgeries. No definite conclusions on the role of energy expenditure can be drawn at this time, and additional mechanisms should be sought to explain the metabolic improvements after bariatric surgery. Changes in Eating Behavior The consumption of diets high in fat has been associated with the development and maintenance of obesity in both humans and rodents [23, 24]. Also obese individuals have a greater propensity to choose high-fat foods, as compared to lean ones [25]. On the other hand, it is known how the eating behaviors change after bariatric surgery. In fact several studies have shown the predilection of lower-fat foods after RYGB [26, 27]. More recently, food choices after SG have been studied in rats [28]. Similarly to what is found after RYGB, in spite of the different anatomic alterations, post-SG rats preferentially choose low-fat and 63 avoid calorie-dense diets [28]. These findings cannot only be explained by the mechanical restriction, as a compensatory choice of more calorie-dense foods to maximize caloric intake would have occurred. Other options to explain such behaviors include postoperative changes of the taste acuity and neural responses to food cues. Two studies have shown enhanced taste acuity and altered hedonic craves for food in post-RYGB patients [29–31]. This has been validated by functional magnetic resonance imaging (fMRI) studies of RYGB patients who presented reduced activation of the mesolimbic reward areas, especially after high-calorie foods [32]. Other possible mechanisms include the aversive symptoms proper of some of the bariatric operations derived by improper food choices. In particular, the development of the uncomfortable symptoms of the dumping syndrome might steer patients away from high caloric carbohydrates. Unfortunately, no scientific evidence on the impact of aversive symptoms and weight loss exists. Occasionally, the aversion to certain foods promotes the development of maladaptive eating behaviors, which ultimately affect the weight loss process. The underlying behavioral and physiological mechanisms of the described phenomenon seem to be complex. However, results from animal models of bariatric surgery indicate that learning processes may play a role as changes in diet selection progress with time in rats after RYGB. ntero-Hormones, Incretins, and Intestinal E Adaptation Gut hormones play a crucial role in regulating appetite, satiety, food intake, systemic metabolism, and insulin secretion. Different degrees of evidence support the physiological roles for ghrelin, CCK, GLP-1, and PYY in GI motility [8]. An important feature of these relationships is that gastric volume, gastric emptying, intestinal-nutrient sensing, and the secretion of these four hormones are linked in negative-­ feedback loops. Changes in GI hormone secretion provide plausible mechanisms for the remarkable therapeutic efficacy of bariatric surgery. Interestingly, similar changes have been seen between patients who have undergone SG and those who have undergone RYGB [8]. After RYGB, the alimentary limb undergoes hyperplasia and hypertrophy, together with increased expression of glucose transporters, increased uptake of glucose into intestinal epithelial cells, and reprogramming of intestinal glucose metabolism to support tissue growth and increased bioenergetic demands. The number of cells producing GLP-1 and GIP within the alimentary limb also increases. Analysis using 2-deoxy-2-[18F] fluoro-D-glucose in rodents and humans show that the alimentary limb becomes a major site for glucose disposal. 64 These changes are likely to contribute to improved glycemic control. In contrast, there is no evidence of GI tract hyperplasia after SG. However, the number and density of cells containing GLP-1 reportedly increase after SG in rodents. Moreover, SG reduces intestinal glucose absorption, potentially contributing to improved glucose tolerance [8]. These findings yet again highlight that RYGB and SG improve glucose homeostasis by different as well as overlapping mechanisms. Entero-Hormones The ingestion of food determines alterations of the gastrointestinal, endocrine, and pancreatic secretions, known as the enteroinsular axis. The main modulators of such mechanism, including GLP-1, GIP, peptide YY, oxyntomodulin, cholecystokinin, and ghrelin, have been found altered after some bariatric surgery procedures (RYGB, BPD-DS, VSG) (Table 5.2). Glucagon-Like Peptide-1 (GLP-1) This is a peptide released by the L cells of the ileum and colon in response to the ingestion of meals. Overall, it is an insulinotropic hormone, and as such, it is responsible for the increase of insulin secretion in response to oral glucose (incretin effect). Additionally it has been linked to stimulate ß(beta)-cell growth, decreasing their apoptosis and, ultimately, increasing their mass in rats [33]. The modulating effect of GLP-1 on postprandial glycemia is also achieved by suppression of glucagon secretion, decreased gastric emptying, and intestinal motility (ileal brake), as well as central nervous system pathways to induce satiety [33, 34]. Overall GLP-1 enhances satiety and reduces food intake. Normally GLP-1 secretion is stimulated by the presence of nutrients in the distal ileum. This is one of the theories to explain the rapid (within days post-procedure) and durable hormonal increase demonstrated after the metabolic procedures with intestinal bypass (RYGB, BPD, BPD-DS) [35–37]. Bypass of a proximal gut (“foregut”) and the increased distal gut (“hindgut”) L-cell nutrient exposure are two potential explanations for the altered gut hormone profile observed after RYGBP. However, the burden of evidence is in favor of the latter. Enhanced GLP-1 responses are also observed after SG, despite the absence of alteration in the route of nutrient delivery. In RYGBP, nutrients rapidly pass through the small gastric pouch, bypassing the majority of the stomach and upper small bowel and directly entering the mid-jejunum. In SG, the removal of the gastric fundus and body results in an unaltered route of nutrient passage through the GI tract [38, 39]. D. R. Funes et al. Both procedures result in accelerated gastric emptying and a rapid entry of undigested food into the jejunum. Consequently, there is enhanced direct contact of nutrients with the apical surface of L cells, interspersed among intestinal epithelial cells, resulting in Ca2-dependent stimulation of GLP-1 secretion into intestinal small blood vessels [40, 41]. Additional mechanisms to explain the GLP-1 increase are related to the inhibition of the GLP-1 degrading enzyme dipeptidyl peptidase-IV (DDP-IV) demonstrated after RYGB and not in type II DM [42]. Once again the evidence is discordant as increased levels of DDP-IV have been reported after BPD [43]. Finally, the role of the GLP-1-induced hunger modulation and decrease in food intake on the weight loss after bariatric operations remain controversial. Evidence for a link between GLP-1 response and weight loss is at best correlative, and a causal relationship has not yet been established. In fact, although the procedures that present the more pronounced weight loss are also the ones determining the highest levels of GLP-1, the increased satiety does not correlate with a significant increase of GLP-1 on longer follow-up studies [44, 45]. We can conclude that although GLP-1 is not the main direct responsible for the weight loss after bariatric operations, it contributes to some weight loss, and it is likely a key contributor to the glycemic homeostasis proper to these procedures. Glucose-Dependent Insulinotropic Polypeptide (GIP) The K cells of the duodenum and proximal jejunum mainly secrete this hormone. Its secretion is enhanced by the presence of nutrients (especially carbohydrates and lipids) in this portion of the intestine. As the name indicates, this is an insulinotropic hormone, although less powerful than GLP-1, determining increased postprandial insulin secretion and pancreatic ß(beta)-cell augmentation [46]. Contrary to GLP-­ 1, GIP has no effect on the intestinal and gastric motility. GIP also affects lipid metabolism by increasing lipogenesis and promoting fat deposition [33]. The function of GIP in diabetic patients is less clear, although consistently demonstrated to be impaired [47]. Due to its site of secretion, GIP has been regarded as one of the hormones possibly involved in the foregut theory. The changes seen in this hormone in animal models of bariatric surgery have not been consistent. Most human studies have reported a decrease in this hormone post-malabsorptive surgery. The effects of bariatric surgery on GIP are discordant, although in general more evidence exists on the decreased levels of this hormone after RYGB and BPD, likely from bypassing the proximal intestine, than the contrary [38, 48]. In contrast, no changes in GLP-1 GIP PYY Oxyntomodulin CCK Ghrelin Origin L cells K cells L cells L cells I cells Oxyntic Satiety ↑ No Δ(delta) ↑ ↑ ↑ ↓ Glycemic control ↑ ↑ ↑ or no Δ(delta) ↑ No Δ No Δ Table 5.2 Characteristics of entero-hormones after bariatric operations GI motility ↓ No Δ(delta) ↓ ↓ ↑ No Δ RYGB ↑ ↓ ↑ ↑ ? ↓ LSG ↑ Unknown ↑ or no Δ(delta) ↑ ↑ or no Δ(delta) ↓↓ LAGB No Δ(delta) No Δ(delta) No Δ(delta) No Δ(delta) Unknown No Δ(delta) BPD ↑ ↓ ↑ ↑ Unknown No Δ(delta) BPD-DS ↑ ↓ ↑ ↑ Unknown ↓↓ 5 Physiological Mechanisms of Bariatric Procedures 65 66 D. R. Funes et al. GIP levels are reported after LAGB [38]. The changes of GIP after LSG remain undetermined. Overall the role of GIP in the mechanism of action of bariatric procedures remains elusive. levels increase after RYGB, but not after LAGB [59]. Because of the overlap in secretion and function, it is difficult to attribute the true value of each one of them in postsurgical weight loss. Peptide Tyrosine Tyrosine (PYY) Cholecystokinin (CCK) PYY is also expressed in the endocrine pancreas, where PYY may have paracrine intraislet actions. Also, PYY is expressed by neurons in the gigantocellular reticular nucleus of the rostral medulla, which have widespread central projections. PYY activates several neuropeptide Y-family receptors (NPYR), including NPY1R (or Y1R), NPY2R, NPY4R, and NPY5R, whereas PYY is selective for NPY2R. NPY2R are expressed throughout the body, including in several brain regions, the GI tract, and vagal afferents [49–51]. PYY may contribute to gastric emptying via the ileal brake mechanism, to the inhibition of eating, and to the control of meal-related glycemia, but the evidence that these are physiological actions remains scarce. Similarly, PYY role in RYGB remains unclear. This modest progress may be due in part to the difficulties of PYY research, including the low threshold for eliciting illness with PYY infusions, the lack of NPY2R antagonists for human use, and the possibility of neuropod PYY signaling. After RYGB, plasma levels of PYY increase modestly (20%) in the fasting state and by 3.5-­ fold in the postprandial state [51]. Similarly, postprandial levels of PYY increase 1 year following VSG. Animal studies support a prominent role for PYY in mediating bariatric weight loss, as postsurgical weight loss is lower in PYY gene knockout as compared with wild-type mice, and infusion of anti-PYY antibodies increases food intake in postbypass rats. Thus, enhanced PYY secretion may contribute to weight loss after RYGB [52–55]. The key role of PYY in the post-bariatric surgery weight loss is supported by encouraging evidence in animal models but needs further dislucidation in human studies which present a challenge given to the current limitations previously discussed. CCK is normally secreted from the duodenum and proximal jejunum in response to nutrients. CCK has been clearly established as a satiation signal in humans and may contribute to the control of meal-related glycemia both indirectly, via its effect on gastric emptying, and directly via control of hepatic glucose production. Pathophysiology of CCK signaling may contribute to overeating, to obesity and T2DM in some patients, and to early satiation after RYGB. Additionally, CCK plays a key role in gallbladder and gastric emptying and exocrine pancreatic secretion. Preclinical studies indicate that CCK is a candidate for obesity pharmacotherapy, especially in combination with other endocrine-based therapies. However, evidence is still unclear on the changes of this hormone after bariatric surgery. Some have shown an increase after LSG, but its overall role in the mechanism of action of these procedures remains undefined [58]. Oxyntomodulin Since its polypeptide structure is similar to GLP-1, oxyntomodulin’s metabolic pathways present several resemblances both in its food-related secretion and degradation process via the enzyme dipeptidyl peptidase-IV (DDP-IV) [56–58]. Similarly to GLP-1, oxyntomodulin reduces gastrointestinal motility and participates in the regulatory mechanism of glucose homeostasis. As seen for the other two hormones secreted by the L cells—GLP-1 and PYY—oxyntomodulin Ghrelin Ghrelin (growth hormone-releasing peptide) is a hormone secreted mainly by the oxyntic glands of the fundus of the stomach and in smaller amounts in the rest of the small bowel. As its name implies, it is involved in the secretion of the growth hormone. This is primarily an orexigenic hormone stimulating directly the hypothalamus. Obese individuals present a decreased suppression of ghrelin after a meal [56, 59, 60]. In addition, ghrelin inhibits insulin secretion by an unknown pathway [61]. It seems that, thanks to this latter property, ghrelin suppresses the insulin-sensitizing hormone adiponectin, negatively affecting the glucose metabolism [62]. Because of these negative effects on the glucose homeostasis, the reduction of ghrelin seen after certain bariatric operations could be beneficial for overall glycemic control [62]. Although most of the biological effects of ghrelin are due to its acylated form, the non-acylated equivalent seems biologically active as well [33]. The challenge in identifying the two forms with different assays might explain some of the discordant findings of ghrelin variation after bariatric operations. In general, although it would be reasonable to speculate that bariatric procedures that do not alter the contact of food with the fundic glands (LAGB, BPD) do not determine significant alteration of ghrelin levels, evidence of the opposite exists [63, 64]. However, if some reports have shown the reduction of ghrelin levels after RYGB, others 5 Physiological Mechanisms of Bariatric Procedures found no changes or even increases of such levels [57, 65, 66]. In randomized trials, ghrelin levels have been found to be permanently lower after LSG than RYGB, likely due to the complete removal of gastric fundus [67]. Also vagal stimulation might affect ghrelin secretion, and vagotomy has been associated with decreased levels [68], although the role of the vagus nerve on the secretion of ghrelin has been disputed by others [69]. Overall, contradicting evidence exists on the role of ghrelin on the weight loss after bariatric surgery, and this hormone likely plays only a marginal role. Mechanisms of Diabetes Resolution The existence of an entero-hormonal mechanism to explain diabetes resolution has been postulated for several years [7]. This is also indirectly proven by the pattern of diabetes resolution after gastric banding that follows the weight loss curve and by the multiple hormonal changes described after gastric bypass [70, 71]. In particular, insulin and leptin levels decrease, whereas GLP-1, GIP, PYY, and ACTH increase even before any significant weight loss [71, 72]. Currently, two main theories exist on the mechanism of diabetes resolution after bariatric surgery: the “foregut” and “hindgut.” Foregut Hypothesis According to this theory, the exclusion of the duodenum from the pathway of the nutrients will prevent the secretion of an unidentified “anti-incretin” substance. In fact, diabetes mellitus (DM) could be due to the overproduction of an “anti-incretin” that determines decreased insulin secretion, insulin resistance, and depletion of the β(beta)-cell mass. When the food bypasses the duodenum, this “anti-incretin” is inhibited. Among the advocates for this theory, Rubino et al. have elegantly demonstrated the resolution of diabetes in rats in which the duodenum was surgically bypassed and excluded [73]. The restoration of duodenal passage in the same group of animals resulted in recurrence of the impaired glucose tolerance state. Others believe that the glucose absorption changes after duodenal bypass. In fact, it has been previously described in a rodent model that both the intestinal morphology and the Na+/glucose cotransporter 1 (SGLT1) function are altered after gastric bypass [74]. In particular, the villous height and crypt depth of the intestinal segments exposed to nutrients are increased, but, unexpectedly, the glucose transport activity is decreased. According to the authors, this could be one of the mechanisms involved in the improvement or resolution of diabetes after duodenal exclusion procedures, such as gastric bypass. Although the process by which duodenal exclusion leads to decreased glucose transport is unclear, some authors have speculated that 67 the interruption of the proximal intestinal regulation of SGLT1 via the sweet taste receptors T1R2 and T1R3 is responsible [74]. Hindgut Hypothesis Additional and/or alternative theories of glucose homeostasis entail the secretions of putative peptides determined by the increase glucose load in the hindgut (“hindgut theory”). According to this second theory, the early presence of undigested food in the distal small bowel stimulates the secretion of “incretin” substances, which, in turn, determines normalization of the glycemia, increases insulin production, and decreases insulin resistance. Although, once again, a single substance has not been identified, GIP and GLP-1 remain the most promising putative candidates. Initially increased GLP-1 and GIP cannot account for improved glucose tolerance, but as glucose normalizes, the action of especially GIP on insulin secretion might be restored [8]. Neuroendocrine Mechanism Experimental models highlight a complex interplay of hormonal and neural pathways that converge and possibly interact at various levels of the gut-brain axis to regulate energy balance. Bariatric surgery procedures, including Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG), influence body weight and glucose regulation via central neural circuits that are recruited by vagally mediated pathways following activation of stretch or chemoreceptors in the stomach. Gut hormones are able to exert their effects on central pathways by either acting locally on vagal endings or via the circulation [75]. Vagus Nerve The activation of central neural circuits, downstream of vagal afferent signaling, was recently suggested to be involved in mediating satiety and glycemic control in a mouse model of RYGB. These data implicates vagal endings in sensing the elevation in gastric distension and nutrients in the roux limb to activate an anorexigenic pathway involving the nucleus tractus solitarius (NTS), lateral parabrachial nucleus, and central nucleus of the amygdala [75]. The activation of this pathway in the immediate period following RYGB is likely to be responsible for the dramatic reduction in food intake after RYGB and may also contribute to the consumption of smaller and slower meals. In addition, there is evidence supporting the reorganization of hindbrain feeding circuits following RYGB and SG. Experimental models of SG indicate 68 that vagal mechanisms contribute to the efficacy of the procedure and that there is a lower threshold for activation of neurons in the NTS in response to a nutrient stimulus, as demonstrated by elevated levels of Fos protein. We have also recently generated data showing increased neural activation, under fasting conditions, within the same vagal circuits. Furthermore, the extent to which vagal mechanisms are central to the reduction in appetite (and weight loss) induced by SG also remains unclear. There is no evidence of the benefits of vagotomy on the postsurgical weight loss. Several trials on LAGB and RYGB have shown no benefits on weight loss by adding a vagotomy [69, 76–79]. Nevertheless, vagally mediated mechanisms seem to have a crucial role in the neuroendocrine mechanisms of RYGB and SG. Bile Acids (BA) Bile acids are synthesized from cholesterol in the liver. Ingestion of food causes bile acid secretion from the gallbladder through the common bile duct to the duodenum. Upon reaching the ileum, bile acids are transported by specific transport proteins to the portal circulation for recycling back to the liver. The two primary bile acids produced by the liver in humans are cholic acid (CA) and chenodeoxycholic acid (CDCA). Bile acids undergo chemical modification through conjugation in the liver and dehydroxylation by gut bacteria [80]. Bile acids also function as a ligand for a specific nuclear transcription factor, the farnesoid X receptor (FXR), which forms a heterodimeric complex with retinoic X receptor-𝛼 (RXR-𝛼) that binds to an inverted repeat sequence in gene promoters. Bile acids not only function in lipid absorption in the gut but also appear to be part of a broader physiological response to ingested nutrients that also involves glucose metabolism [81]. This is consistent with the anabolic need to store fatty acids as triglycerides, which requires a glycerol-­3-­ phosphate backbone that is derived from glucose. The effects of bile acids on the glucose metabolism might be mediated by the activation of the L cells via bile acid-TGR5 (G protein-­ coupled bile acid receptor) and FXR signaling [86]. When recognized by TGR5 and FXR-α receptor in the liver, bile acid induces liver glycogen synthesis, inhibits gluconeogenesis, ameliorates body’s insulin sensitivity, and controls glucose metabolism. FXR−/− mice exhibit peripheral insulin resistance, reduced glucose disposal, and decreased adipose tissue and skeletal muscle insulin signaling, and, conversely, activation of FXR by the agonist GW4064 in insulin-­resistant ob/ob mice reduced hyperinsulinemia and improved glucose tolerance. The hindgut hypothesis is based on the premise that inappropriate delivery of ingested nutrients and/or digestive juices to more distal regions of the small intestine induces a putative molecular mediator that ameliorates T2D D. R. Funes et al. [82]. Bile acids have been implicated as key molecules in this hypothesis. Recent work in both clinical studies and animal models supports a key role for bile acids. Systemic BA levels are elevated in patients following RYGB, suggesting an increase in BA signaling after RYGB. SG has been shown to modify the expression of certain hepatic genes involved in the metabolism of bile acid [83–85]. Furthermore, recently FXR has been shown to be required for the antidiabetic effect of SG in mice [28]. Also SG resulting in resolution of T2D seems to contradict the hindgut hypothesis, inappropriate delivery of nutrients and digestive components to the distal intestine hypothesis. However, gastric transit is substantially increased in SG, expediting delivery through the duodenum into the distal intestine. The binding of bile acids with the nuclear receptor FXR (farnesoid X receptor) has been associated with positive alterations of the feeding behavior (repression of rebound hyperphagia), improved glucose tolerance, and likely alteration of the gut flora in post-vertical sleeve gastrectomy mice, as opposed to post-VSG FXR knockout counterpart [87]. Further delineation of the molecular mechanisms underlying these beneficial effects could provide target for novel, less invasive, and efficient treatments. Gastrointestinal Microflora The composition of the gastrointestinal microflora established during the first year of life influenced by a variety of environmental and metabolic factors is relatively stable during adulthood. However, the adult colon has rich microbial diversity resulting from the estimated 1000–36,000 different bacterial species contained within its lumen [103]. This diverse bacterial population contains perhaps 100 times more genes than the human genome [104]. The coexistence of the intestinal microbiota is essential for several host functions, such as vitamin synthesis. Recently additional links between gut flora and the metabolism have been discovered. Instrumental in this process is the fact that both mouse and human microbiota are prevalently populated by the same bacterial species: Bacteroidetes and Firmicutes. Comparisons of the distal gut microbiota in genetically obese mice and their lean littermates have revealed that changes in the relative abundance of the two dominant bacterial divisions, the Bacteroidetes and Firmicutes, are associated with the level of adiposity [105–107]. Specifically, obese mice have a significantly higher level of Firmicutes and lower levels of Bacteroidetes compared with their lean counterparts [108]. Similar results have been established in humans [107]. Furthermore, biochemical analyses have indicated that such shifts in microbial community structure are associated with an increased efficiency in energy harvest in obese individuals 5 Physiological Mechanisms of Bariatric Procedures from a given caloric load; these findings suggest that the gut microbiota may be a significant contributor to an individual’s energy balance. It has been well documented that weight loss is of great benefit in obese patients with type II diabetes mellitus (T2DM), often eliminating the need for pharmacologic intervention to treat insulin resistance [109, 110]. It has also been established that diet-induced weight loss in humans has a marked effect on gut microbial ecology—shifting the gut microbial community composition toward that seen in lean individuals [107]. Intriguingly, experimental alteration of intestinal flora in genetically obese mice results in weight loss independent of improvement of glycemia [111]. The division-wide change in microbial ecology that has been associated with obesity suggests that the obese gut microbiota may play an important role in the morbidity associated with obesity, and its modification might be responsible for the resolution of some comorbidities. Changes in the composition of the gut microflora after RYGB are a potential contributor to both weight loss and comorbidity resolution. However, this mechanism has received little attention. Zhang et al. demonstrated that the Firmicutes were decreased in three gastric bypass patients compared to normal-weight and obese individuals [112]. Meanwhile, Woodard et al. directly manipulated the gastrointestinal microbiota using a Lactobacillus probiotic agent following gastric bypass [113]. They showed that the probiotic group had greater weight loss than matched controls. In a mouse model, SG is associated with changes in the gut microbiome at 1 week and persists 1 month following surgery. The identified increases in members of the Enterobacteriaceae, Enterococcaceae, and Porphyromonadaceae families correlate with reduced weight. Enterobacteriaceae, within Proteobacteria, has been observed to increase following SG and RYGB in mice by others as well. This parallels observations in humans following RYGB. These experiments suggest that the gastrointestinal microbiota may play a significant role in human energy homeostasis. Although it is clear that microbiota play important roles in many aspects of energy metabolism, further work is needed to characterize and identify the metabolic contribution of gut microbial changes associated with SG. Adipose Tissue The excessive peripheral deposition of fat has been associated with peripheral and hepatic insulin resistance [88]. Furthermore, it is well known how the visceral fat constitutes a true hormone-producing substrate. Consequently, obese patients present increased levels of pro-inflammatory cytokines such as TNF, interleukin-6, and leptin and reduced levels of anti-inflammatory hormones such as adiponectin [89]. 69 The impact of bariatric surgery on the inflammatory markers, specifically which inflammatory markers are closely associated with changes in obesity and improvements in insulin sensitivity, needs further delineation. The endocrine role of the adipose fat has been well established [90]. Among the multiple adipokines described, omentin-1 has been more recently described as an important modulator of insulin sensitivity [91, 92]. Plasma omentin-1 levels and its adipose tissue gene expression are markedly decreased in obese individuals [92]. Plasma omentin-1 levels are positively correlated with both adiponectin and HDL levels and negatively with insulin resistance [92]. The omentin genes are located in the same chromosomal region associated with the development of type II diabetes [93, 94]. Leptin A paramount contribution for the comprehension of the regulatory mechanisms between food intake and energy regulation has been the discovery of the adipokine leptin. As it stands now, levels of circulating leptin result from the contribution of two major organs, the white adipose tissue and the gastric mucosa. This hormone crosses the blood-brain barrier, and its main sites of action are the hypothalamic cells where it plays fundamental roles in the control of appetite and in the regulation of energy expenditure. At first it was considered a hormone specific to the white adipose tissue; however, recently it was found expressed by other tissues. Among these, the gastric mucosa has been demonstrated to secrete large amounts of leptin. Secretion of leptin by the gastric chief cells was found to be an exocrine secretion. While secretion of leptin by the white adipose tissue is constitutive, secretion by the gastric cells is a regulated one responding very rapidly to secretory stimuli such as food intake [95]. In general, decreased levels of leptin have been associated with increased hunger [96]. Some authors suggested a direct link between leptin and inhibition of lipogenesis and increased lipolysis. In fact, obese individuals have an increased baseline concentration of leptin, and the levels decrease after weight loss [97]. Since the reduction of leptin also leads to a reduction in energy expenditure, the maintenance of weight loss simply through diet becomes challenging [51]. The reduction of leptin has been reported in all the bariatric procedures (RYGB, LSG, LAGB), and it has been linked directly with weight loss [52]. Interestingly, post-RYGB patients who remain obese present a decreased level of leptin, suggesting mechanisms other than weight loss to explain the postoperative changes [98]. Further studies regarding the effects of bariatric surgery in the gastric mucosal leptin secretion are needed to determine the extent of its contribution to the well-­ established physiological mechanisms of bariatric surgery. 70 D. R. Funes et al. Table 5.3 Changes of adipocytokine after bariatric operations Obese Post RYGB Leptin ↑ ↓ Adiponectin ↓ ↑ Omentin ↓ ↑ Adiponectin Adiponectin is also produced by the adipose tissue, and it is related to insulin sensitivity and fatty acid oxidation [99]. Contrary to leptin, adiponectin levels are decreased in obese patients and increase with weight loss [100]. Low adiponectin levels are associated with insulin resistance and coronary artery disease [101]. After RYGB, the levels of adiponectin increase and correlate with the improved insulin sensitivity measured by HOMA-IR [98]. Furthermore, lower ­preoperative levels of adiponectin have been linked to greater increase in postoperative levels and increased weight loss, maybe because of enhanced fatty acid oxidation into the muscle [98]. The adiponectin-related decrease in TNFα(alpha) has been advocated as a potential mechanism to decrease monocyte adhesion to the endothelial cells [102]. The reason for the changes of the mentioned cytokines after RYGB seems to be related mainly, but not exclusively, to the weight loss, as it has been similar for other bariatric procedures and for calorie-controlled diets [51] (Table 5.3). β(Beta)-Cell Changes Besides the previously mentioned gastrointestinal hormones, residual ß(beta)-cell function has been implicated as a determinant in the glycemic control after bariatric operations [114]. In fact, the rate of remission of diabetes has been linked to the patient-specific characteristics of the diabetes itself. Shorter diabetes duration, lesser degree of β(beta)-cell dysfunction (C-peptide positive), and lesser or no insulin requirements have been linked to higher chance of diabetes remission after surgery [114, 115]. Also it has been shown how, on one hand, RYGB results in an improvement of insulin sensitivity proportionally to the weight loss, but on the other hand, β(beta)-cell glucose sensitivity increases independently from it [116]. To further validate the importance of the residual ß(beta)-cell function for the remission of diabetes, some studies have shown the lack of significant benefit of RYGB in glycemic control of type I DM, in spite of similar changes of GLP-1 and weight loss as in type II DM patients [114]. It is important to note that some, and probably less convincing, evidence exists of type I DM amelioration after RYGB. In fact, in a small series of three patients, a significant and durable (8 years) improvement in glycemic control was demonstrated, suggesting other mechanism other than residual β(beta)-cell function [117]. However, the increase of GLP-1 after type I DM, although comparable with a similar increase in type II DM patients, does not determine suppression of glucagon secretion, but rather an increase [114]. This unexplained phenomenon, once again, suggests additional factors responsible for glycemic control after bariatric operations besides the degree of β(beta)-cell function. However, more recent animal studies have demonstrated pancreatic islet cell histopathological changes following RYGB showing reduced islets interstitial fibrosis, as well as regeneration and hypertrophy of beta cell mass. These findings suggest that RYGB may have a role in promoting islet regeneration. Notch signaling is an evolutionary conserved pathway of cell-cell communication and cell-fate determination during embryonic development and tissue homeostasis. The results from previously mentioned animal studies indicate a potential role of this signaling pathway in regulating the pancreatic islet regeneration following RYGB. End-Organ Changes Increased Insulin Sensitivity The beneficiary effects of bariatric surgery are evident on both the insulin secretion and the improvement of insulin sensitivity. In general, weight loss determines increases in peripheral insulin sensitivity, but this is not the only mechanism after bariatric surgery. The most convincing evidence of increased peripheral insulin sensitivity derives from the studies on BPD. Mari et al., in fact, using the hyperinsulinemic-euglycemic clamp methodology, demonstrated significant improvement of the insulin sensitivity within the day of the procedure [118]. The data for RYGB is, instead, discordant [119–123]. No significant changes have been shown in the LAGB and LSG studies [120]. ardiac Response to Obesity and Bariatric C Surgery The “thrifty gene hypothesis” of James Neel postulates that obesity and type 2 diabetes were an evolutionary advantage in times of starvation [124]. In modern times, the ability to continually store energy has become a major disadvantage. Obesity and the development of associated complications are multifactorial and dictated by the individual exposures which include the ones that enter the body from the environment as well as those generated in excess within the body by inflammation, oxidative stress, and metabolic dysregulation. There are specific mechanisms that the body uses in an attempt to reestablish homeostasis [125]. In obesity, the body’s initial response to excess energy is to partition it into adipose tissue 5 Physiological Mechanisms of Bariatric Procedures to reduce the metabolic burden on its major organs such as the heart. However, when the storage capacity of adipose tissue is exceeded, energy is stored ectopically as fat in vital organs creating a lipotoxic environment in non-adipose tissue. In obesity, through environmental and subsequently intrinsic factors, there is an increased hemodynamic, neurohormonal, and metabolic load. There is strong evidence that the deposition and also the utilization of fatty acids as source of energy by the cardiac tissue are associated with increased myocardial oxygen consumption and decreased mechanical efficiency. Such ­ studies have helped to establish obesity as an independent predictor of increased myocardia oxygen consumption and increased cardiovascular risk [125, 126]. Just until recently it is widely recognized that bariatric surgery improves cardiac function in obese patients and also reduces the risk of cardiovascular disease. The principal mechanisms involved in the benefits of bariatric surgery, predominantly RYGB and SG, occur at a micro cellular level in the endoplasmic reticulum (ER). The ER is responsible for balancing the nutritional status of the cell, homeostasis that is vital for protein synthesis, energy storage, and continued nutrient sensing. When supplied with excess substrate, ER undergoes stress, widely associated with cardiac dysfunction and disease. In bariatric surgery, the most studied procedure in this regard is RYGB, which has shown to reduce the metabolic load and stress to the heart making it plausible that the beneficial cardiovascular effects may be mediated by the reduction of cardiac ER stress or load [127]. Gastrointestinal-Renal Axis There is growing evidence strengthening the rationale to further explore the hypothesis of the existence of a direct communication between the gastrointestinal (GI) tract and the kidneys. This theory is based on the fact that GI hormones and peptides have shown to regulate the autocrine function of renal hormones, affecting renal function and including sodium excretion. GLP-1 is the GI hormone that has been most widely studied in this regard, demonstrating a direct and indirect natriuretic effect by targeting early tubular sodium reabsorption [128]. Moreover, along with reference to emerging data demonstrating that GLP-1 can exert direct anti-inflammatory and antioxidant effects in the kidney, we can now explain the link between the beneficial effects of bariatric surgery on hypertension resolution and kidney function improvement. Animal models provide one of the most important pieces of evidence linking the physiological mechanisms of bariatric surgery and the GI-renal axis; these are the effects of glucagon-like peptide 1 (GLP-1) and GLP-1 receptor agonists (GLP-1RAs) on renal hemodynamics. Several vascular and tubular factors result in a net reduction 71 in afferent renal arteriolar resistance, a net increase in efferent renal arteriolar resistance and/or a reduction in hydraulic pressure in Bowman space, and thereby an increase in glomerular hydraulic pressure and single-nephron glomerular filtration rate [128, 129]. GLP-1RAs are associated with direct GLP-1R-mediated and, at least in part, nitric oxide-­ dependent vasodilation of the afferent renal arteriole, as well as indirect inhibition of vascular and tubular factors that are putative mediators of glomerular hyperfiltration, which is well known to be the primary event resulting in chronic kidney injury [128, 130]. In an experimental model, the integrated effect of incretin-­ based therapy on renal hemodynamics seems to be the result of direct vasodilative actions and inhibition of pathways of glomerular hyperfiltration, which may lead us to believe that the changes in GLP-1 following bariatric surgery may result in a similar effect on renal hemodynamics [128]. ariatric Surgery and the Control of Blood B Pressure Through the GI-Renal Axis The kidney plays a vital role in the regulation of sodium balance and blood pressure. However, the gastrointestinal (GI) tract, which is the organ first exposed to components of food, has taste receptors and sensors for electrolytes including sodium [130]. Therefore, in addition to the kidney, there is increasing realization of the importance of the GI tract in the regulation of sodium balance and consequently on blood pressure level. Excessive sodium in the body, as a consequence of increased dietary intake and/or impaired excretion, is the most common risk factor for hypertension. In addition to renal regulation of sodium homeostasis, which has been widely described, gastrointestinal absorption and its control via GI hormones play a critical role in the control of blood pressure. Most of the electrolytes including sodium are absorbed in the small (95%) and large (4%) intestine. The intestinal absorption of fluid by GI epithelial cells occurs via active transport of (NaCl) sodium chloride. NaCl absorption occurs from the small intestine to the distal colon. The GI-derived hormones can be grouped into three classes: GI hormones, pancreatic hormones, and GI neuropeptides. According to their ability to affect sodium excretion, we can further classify these hormones and neuropeptides into two groups: a group that increases and another group that decreases sodium excretion. Of these hormones, ghrelin and GLP-1 have the strongest evidence implicating them with the beneficial effects of bariatric surgery [130, 131]. In animal models, ghrelin exerts its effect on distal nephron-promoting diuresis and renal nitric oxide production. Additionally, ghrelin also stimulates nephron-dependent sodium reabsorption. Ghrelin’s direct effect in the function of renal cells consists in the reduction of mitochondrial 72 membrane potential, and mitochondrial-derived reactive oxygen species (ROS) ameliorates angiotensin II-induced cell senescent in the renal proximal tubule cells. GLP-1 inhibits sodium uptake and facilitates natriuresis, and an animal model demonstrates the effect of GLP-1 in the brush border of proximal tubules inhibiting the sodium-hydrogen antiporter 3 (NHE3) activity and sodium reabsorption in RPT cells. Both long-term and short-term studies have shown that the blood pressure decreases in adults who underwent RYGB and SG. Compared with RYGB, sleeve gastrectomy is associated with better early remission rates for hypertension. Overall evidence suggest that this may be related to the ability of bariatric surgery to increase the plasma levels of natriuretic enterokines such as GLP-1 [130–132]. Conclusion Although the mechanism of action of the different bariatric operations is not completely understood, multiple factors seem to play a role. The weight loss seems only in part due to purely restrictive mechanisms. Hormonal changes stimulate anorexigenic pathways in the brain. Furthermore, the role of bile salts and the gastrointestinal microflora needs further elucidation. Similarly the resolution of diabetes appears to be a multifactorial process. It is likely that two of the major early contributors are the increased hepatic insulin sensitivity due to caloric restriction and the improved ß(beta)-cell function secondary to increased entero-hormones caused by altered exposure of the distal small intestine to nutrients. Later changes of the glucose homeostasis are likely due to weight loss-induced improvement of peripheral skeletal muscle insulin sensitivity. Question Section 1. Which one of the following gut hormones increases after RYGB? A. GLP-1 B. Ghrelin C. GIP D. A + C E. A + B 2. Which one of the following statements is/are TRUE about leptin? A. Leptin is inversely associated with hunger. B. Leptin increases lipolysis and decreases lipogenesis. C. Leptin decreases after bariatric surgery. D. Leptin is directly related to energy expenditure. E. All of the above. D. R. Funes et al. References 1. Hales CM, Carroll MD, Fryar CD, Ogden CL. Prevalence of obesity among adults and youth, 2015–2017.NCHS Data Brief. 2017;(288):1–8. 2. Finkelstein EA, Khavjou OA, Thompson H, Trogdon JG, Pan L, Sherry B, Dietz W. Obesity and severe obesity forecasts through 2030. Am J Prev Med. 2012;42(6):563–70. 3. Sjöström L, Lindroos A-K, Peltonen M, Torgerson J, Bouchard C, Carlsson B, Dahlgren S, Larsson B, Narbro K, Sjöström CD, Sullivan M, Wedel H. Lifestyle, diabetes, and cardiovascular risk factors 10 years after bariatric surgery. N Engl J Med. 2004;351(26):2683–93. 4. Sumithran P, Prendergast LA, Delbridge E, Purcell K, Shulkes A, Kriketos A, Proietto J. Long-term persistence of hormonal adaptations to weight loss. N Engl J Med. 2011;365(17):1597–604. 5. Schwartz MW, Woods SC, Porte D, Seeley RJ, Baskin DG. Central nervous system control of food intake. Nature. 2000;404(6778):661–71. 6. Pories WJ, Swanson MS, MacDonald KG, Long SB, Morris PG, Brown BM, Barakat HA, deRamon RA, Israel G, Dolezal JM. Who would have thought it? An operation proves to be the most effective therapy for adult-onset diabetes mellitus. Ann Surg. 1995;222(3):339–50. 7. Mason EE. Ileal [correction of ilial] transposition and enteroglucagon/GLP-1 in obesity (and diabetic?) surgery. Obes Surg. 1999;9(3):223–8. 8. Batterham RL, Cummings DE. Mechanisms of diabetes improvement following bariatric/metabolic surgery. Diabetes Care. 2016;39(6):893–901. 9. Kumar R, Lieske JC, Collazo-Clavell ML, Sarr MG, Olson ER, Vrtiska TJ, Bergstralh EJ, Li X. Fat malabsorption and increased intestinal oxalate absorption are common after Roux-en-Y gastric bypass surgery. Surgery. 2011;149(5):654–61. 10. Odstrcil EA, Martinez JG, Santa Ana CA, Xue B, Schneider RE, Steffer KJ, Porter JL, Asplin J, Kuhn JA, Fordtran JS. The contribution of malabsorption to the reduction in net energy absorption after long-limb Roux-en-Y gastric bypass. Am J Clin Nutr. 2010;92(4):704–13. 11. Buchwald H, Avidor Y, Braunwald E, Jensen MD, Pories W, Fahrbach K, Schoelles K. Bariatric surgery: a systematic review and meta-analysis. JAMA. 2004;292(14):1724–37. 12. Kelley DE, Wing R, Buonocore C, Sturis J, Polonsky K, Fitzsimmons M. Relative effects of calorie restriction and weight loss in noninsulin-dependent diabetes mellitus. J Clin Endocrinol Metab. 1993;77(5):1287–93. 13. Kirk E, Reeds DN, Finck BN, Mayurranjan SM, Mayurranjan MS, Patterson BW, Klein S. Dietary fat and carbohydrates differentially alter insulin sensitivity during caloric restriction. Gastroenterology. 2009;136(5):1552–60. 14. Isbell JM, Tamboli RA, Hansen EN, Saliba J, Dunn JP, Phillips SE, Marks-Shulman PA, Abumrad NN. The importance of caloric restriction in the early improvements in insulin sensitivity after Roux-en-Y gastric bypass surgery. Diabetes Care. 2010;33(7):1438–42. 15. Pournaras DJ, Osborne A, Hawkins SC, Vincent RP, Mahon D, Ewings P, Ghatei MA, Bloom SR, Welbourn R, le Roux CW. Remission of type 2 diabetes after gastric bypass and banding: mechanisms and 2 year outcomes. Ann Surg. 2010;252(6):966–71. 16. Oliván B, Teixeira J, Bose M, Bawa B, Chang T, Summe H, Lee H, Laferrère B. Effect of weight loss by diet or gastric bypass surgery on peptide YY3-36 levels. Ann Surg. 2009;249(6):948–53. 17. Schauer PR, Kashyap SR, Wolski K, Brethauer SA, Kirwan JP, Pothier CE, Thomas S, Abood B, Nissen SE, Bhatt DL. Bariatric surgery versus intensive medical therapy in obese patients with diabetes. N Engl J Med. 2012;366(17):1567–76. 5 Physiological Mechanisms of Bariatric Procedures 18. Lips MA, de Groot GH, van Klinken JB, Aarts E, Berends FJ, Janssen IM, Van Ramshorst B, Van Wagensveld BA, Swank DJ, Van Dielen F, Willems van Dijk K, Pijl H. Calorie restriction is a major determinant of the short-term metabolic effects of gastric bypass surgery in obese type 2 diabetic patients. Clin Endocrinol. 2014;80:834–42. 19. Schwartz A, Doucet E. Relative changes in resting energy expenditure during weight loss: a systematic review. Obes Rev. 2010;11(7):531–47. 20. Benedetti G, Mingrone G, Marcoccia S, Benedetti M, Giancaterini A, Greco AV, Castagneto M, Gasbarrini G. Body composition and energy expenditure after weight loss following bariatric surgery. J Am Coll Nutr. 2000;19(2):270–4. 21. Carrasco F, Papapietro K, Csendes A, Salazar G, Echenique C, Lisboa C, Díaz E, Rojas J. Changes in resting energy expenditure and body composition after weight loss following Roux-en-Y gastric bypass. Obes Surg. 2007;17(5):608–16. 22. Werling M, Olbers T, Fändriks L, Bueter M, Lönroth H, Stenlöf K, le Roux CW. Increased postprandial energy expenditure may explain superior long term weight loss after Roux-en-Y gastric bypass compared to vertical banded gastroplasty. PLoS One. 2013;8(4):e60280. 23. Winzell MS, Ahrén B. The high-fat diet-fed mouse: a model for studying mechanisms and treatment of impaired glucose tolerance and type 2 diabetes. Diabetes. 2004;53(Suppl 3):S215–9. 24. Hill JO, Melanson EL, Wyatt HT. Dietary fat intake and regulation of energy balance: implications for obesity. J Nutr. 2000;130(2S Suppl):284S–8. 25. Drewnowski A, Kurth C, Holden-Wiltse J, Saari J. Food preferences in human obesity: carbohydrates versus fats. Appetite. 1992;18(3):207–21. 26. Thomas JR, Marcus E. High and low fat food selection with reported frequency intolerance following Roux-en-Y gastric bypass. Obes Surg. 2008;18(3):282–7. 27. Ernst B, Thurnheer M, Wilms B, Schultes B. Differential changes in dietary habits after gastric bypass versus gastric banding operations. Obes Surg. 2009;19(3):274–80. 28. Wilson-Pérez HE, Chambers AP, Sandoval DA, Stefater MA, Woods SC, Benoit SC, Seeley RJ. The effect of vertical sleeve gastrectomy on food choice in rats. Int J Obes. 2013;37(2): 288–95. 29. Delin CR, Watts JM, Saebel JL, Anderson PG. Eating behavior and the experience of hunger following gastric bypass surgery for morbid obesity. Obes Surg. 1997;7(5):405–13. 30. Burge JC, Schaumburg JZ, Choban PS, DiSilvestro RA, Flancbaum L. Changes in patients’ taste acuity after Roux-en-Y gastric bypass for clinically severe obesity. J Am Diet Assoc. 1995;95(6):666–70. 31. Scruggs D, Buffington C, Cowan G. Taste acuity of the morbidly obese before and after gastric bypass surgery. Obes Surg. 1994;4(1):24–8. 32. Ochner CN, Kwok Y, Conceição E, Pantazatos SP, Puma LM, Carnell S, Teixeira J, Hirsch J, Geliebter A. Selective reduction in neural responses to high calorie foods following gastric bypass surgery. Ann Surg. 2011;253(3):502–7. 33. Ionut V, Burch M, Youdim A, Bergman RN. Gastrointestinal hormones and bariatric surgery-induced weight loss. Obesity (Silver Spring). 2013;21(6):1093–103. 34. Vetter ML, Ritter S, Wadden TA, Sarwer DB. Comparison of bariatric surgical procedures for diabetes remission: efficacy and mechanisms. Diabetes Spectr. 2012;25(4):200–10. 35. Jørgensen NB, Jacobsen SH, Dirksen C, Bojsen-Møller KN, Naver L, Hvolris L, Clausen TR, Wulff BS, Worm D, Lindqvist Hansen D, Madsbad S, Holst JJ. Acute and long-term effects of Roux-en-Y gastric bypass on glucose metabolism in subjects with type 2 diabetes and normal glucose tolerance. Am J Physiol Endocrinol Metab. 2012;303(1):E122–31. 73 36. le Roux CW, Welbourn R, Werling M, Osborne A, Kokkinos A, Laurenius A, Lönroth H, Fändriks L, Ghatei MA, Bloom SR, Olbers T. Gut hormones as mediators of appetite and weight loss after Roux-en-Y gastric bypass. Ann Surg. 2007;246(5):780–5. 37. Guidone C, Manco M, Valera-Mora E, Iaconelli A, Gniuli D, Mari A, Nanni G, Castagneto M, Calvani M, Mingrone G. Mechanisms of recovery from type 2 diabetes after malabsorptive bariatric surgery. Diabetes. 2006;55(7):2025–31. 38. Korner J, Bessler M, Inabnet W, Taveras C, Holst JJ. Exaggerated glucagon-like peptide-1 and blunted glucose-dependent insulinotropic peptide secretion are associated with Roux-en-Y gastric bypass but not adjustable gastric banding. Surg Obes Relat Dis. 2007;3(6):597–601. 39. McLaughlin T, Peck M, Holst J, Deacon C. Reversible hyperinsulinemic hypoglycemia after gastric bypass: a consequence of altered nutrient delivery. J Clin Endocrinol Metab. 2010;95(4):1851–5. 40. Goldfine AB, Mun EC, Devine E, Bernier R, Baz-Hecht M, Jones DB, Schneider BE, Holst JJ, Patti ME. Patients with neuroglycopenia after gastric bypass surgery have exaggerated incretin and insulin secretory responses to a mixed meal. J Clin Endocrinol Metab. 2007;92(12):4678–85. 41. Romero F, Nicolau J, Flores L, Casamitjana R, Ibarzabal A, Lacy A, Vidal J. Comparable early changes in gastrointestinal hormones after sleeve gastrectomy and Roux-En-Y gastric bypass surgery for morbidly obese type 2 diabetic subjects. Surg Endosc. 2012;26(8):2231–9. 42. Alam ML, Van der Schueren BJ, Ahren B, Wang GC, Swerdlow NJ, Arias S, Bose M, Gorroochurn P, Teixeira J, McGinty J, Laferrère B. Gastric bypass surgery, but not caloric restriction, decreases dipeptidyl peptidase-4 activity in obese patients with type 2 diabetes. Diabetes Obes Metab. 2011;13(4):378–81. 43. Lugari R, Dei Cas A, Ugolotti D, Barilli AL, Camellini C, Ganzerla GC, Luciani A, Salerni B, Mittenperger F, Nodari S, Gnudi A, Zandomeneghi R. Glucagon-like peptide 1 (GLP-1) secretion and plasma dipeptidyl peptidase IV (DPP-IV) activity in morbidly obese patients undergoing biliopancreatic diversion. Horm Metab Res. 2004;36(2):111–5. 44. Falkén Y, Hellström PM, Holst JJ, Näslund E. Changes in glucose homeostasis after Roux-en-Y gastric bypass surgery for obesity at day three, two months, and one year after surgery: role of gut peptides. J Clin Endocrinol Metab. 2011;96(7):2227–35. 45. Pournaras DJ, Osborne A, Hawkins SC, Mahon D, Ghatei MA, Bloom SR, Welbourn R, le Roux CW. The gut hormone response following Roux-en-Y gastric bypass: cross-sectional and prospective study. Obes Surg. 2010;20(1):56–60. 46. Meier JJ, Nauck MA, Schmidt WE, Gallwitz B. Gastric inhibitory polypeptide: the neglected incretin revisited. Regul Pept. 2002;107(1–3):1–13. 47. Vollmer K, Holst JJ, Baller B, Ellrichmann M, Nauck MA, Schmidt WE, Meier JJ. Predictors of incretin concentrations in subjects with normal, impaired, and diabetic glucose tolerance. Diabetes. 2008;57(3):678–87. 48. Clements RH, Gonzalez QH, Long CI, Wittert G, Laws HL. Hormonal changes after Roux-en Y gastric bypass for morbid obesity and the control of type-II diabetes mellitus. Am Surg. 2004;70(1):1–4.. discussion 4–5 49. Steinert RE, Feinle-Bisset C, Asarian L, Horowitz M, Beglinger C, Geary N. Ghrelin, CCK, GLP-1, and PYY(3-36): secretory controls and physiological roles in eating and glycemia in health, obesity, and after RYGB. Physiol Rev. 2017;97(1):411–63. 50. Meek CL, Lewis HB, Reimann F, Gribble FM, Park AJ. The effect of bariatric surgery on gastrointestinal and pancreatic peptide hormones. Peptides. 2016;77:28–37. 51. Batterham RL, Cohen MA, Ellis SM, Le Roux CW, Withers DJ, Frost GS, Ghatei MA, Bloom SR. Inhibition of food intake in obese subjects by peptide YY3-36. N Engl J Med. 2003;349(10):941–8. 74 52. Arble DM, Sandoval DA, Seeley RJ. Mechanisms underlying weight loss and metabolic improvements in rodent models of bariatric surgery. Diabetologia. 2015;58(2):211–20. 53. Korner J, Leibel RL. To eat or not to eat – how the gut talks to the brain. N Engl J Med. 2003;349(10):926–8. 54. Korner J, Inabnet W, Conwell IM, Taveras C, Daud A, Olivero-­ Rivera L, Restuccia NL, Bessler M. Differential effects of gastric bypass and banding on circulating gut hormone and leptin levels. Obesity (Silver Spring). 2006;14(9):1553–61. 55. Bose M, Machineni S, Oliván B, Teixeira J, McGinty JJ, Bawa B, Koshy N, Colarusso A, Laferrère B. Superior appetite hormone profile after equivalent weight loss by gastric bypass compared to gastric banding. Obesity (Silver Spring). 2010;18(6):1085–91. 56. Möhlig M, Spranger J, Otto B, Ristow M, Tschöp M, Pfeiffer AFH. Euglycemic hyperinsulinemia, but not lipid infusion, decreases circulating ghrelin levels in humans. J Endocrinol Investig. 2002;25(11):RC36–8. 57. Cummings DE, Overduin J, Foster-Schubert KE. Gastric bypass for obesity: mechanisms of weight loss and diabetes resolution. J Clin Endocrinol Metab. 2004;89(6):2608–15. 58. Karamanakos SN, Vagenas K, Kalfarentzos F, Alexandrides TK. Weight loss, appetite suppression, and changes in fasting and postprandial ghrelin and peptide-YY levels after Roux-en-Y gastric bypass and sleeve gastrectomy: a prospective, double blind study. Ann Surg. 2008;247(3):401–7. 59. Valderas JP, Irribarra V, Boza C, de la Cruz R, Liberona Y, Acosta AM, Yolito M, Maiz A. Medical and surgical treatments for obesity have opposite effects on peptide YY and appetite: a prospective study controlled for weight loss. J Clin Endocrinol Metab. 2010;95(3):1069–75. 60. Akkary E. Bariatric surgery evolution from the malabsorptive to the hormonal era. Obes Surg. 2012;22(5):827–31. 61. Laferrère B, Swerdlow N, Bawa B, Arias S, Bose M, Oliván B, Teixeira J, McGinty J, Rother KI. Rise of oxyntomodulin in response to oral glucose after gastric bypass surgery in patients with type 2 diabetes. J Clin Endocrinol Metab. 2010;95(8):4072–6. 62. Castañeda TR, Tong J, Datta R, Culler M, Tschöp MH. Ghrelin in the regulation of body weight and metabolism. Front Neuroendocrinol. 2010;31(1):44–60. 63. Dezaki K, Sone H, Koizumi M, Nakata M, Kakei M, Nagai H, Hosoda H, Kangawa K, Yada T. Blockade of pancreatic islet-­ derived ghrelin enhances insulin secretion to prevent high-fat diet-­ induced glucose intolerance. Diabetes. 2006;55(12):3486–93. 64. Cummings DE, Foster-Schubert KE, Overduin J. Ghrelin and energy balance: focus on current controversies. Curr Drug Targets. 2005;6(2):153–69. 65. Frühbeck G, Diez-Caballero A, Gil MJ, Montero I, Gómez-­ Ambrosi J, Salvador J, Cienfuegos JA. The decrease in plasma ghrelin concentrations following bariatric surgery depends on the functional integrity of the fundus. Obes Surg. 2004;14(5):606–12. 66. Pérez-Romero N, Serra A, Granada ML, Rull M, Alastrué A, Navarro-Díaz M, Romero R, Fernández-Llamazares J. Effects of two variants of Roux-en-Y gastric bypass on metabolism behaviour: focus on plasma ghrelin concentrations over a 2-year follow­up. Obes Surg. 2010;20(5):600–9. 67. Cummings DE, Weigle DS, Frayo RS, Breen PA, Ma MK, Dellinger EP, Purnell JQ. Plasma ghrelin levels after diet-­ induced weight loss or gastric bypass surgery. N Engl J Med. 2002;346(21):1623–30. 68. Holdstock C, Engström BE, Ohrvall M, Lind L, Sundbom M, Karlsson FA. Ghrelin and adipose tissue regulatory peptides: effect of gastric bypass surgery in obese humans. J Clin Endocrinol Metab. 2003;88(7):3177–83. 69. Peterli R, Wölnerhanssen B, Peters T, Devaux N, Kern B, Christoffel-Courtin C, Drewe J, von Flüe M, Beglinger C. Improvement in glucose metabolism after bariatric surgery: D. R. Funes et al. comparison of laparoscopic Roux-en-Y gastric bypass and laparoscopic sleeve gastrectomy: a prospective randomized trial. Ann Surg. 2009;250(2):234–41. 70. Sundbom M, Holdstock C, Engström BE, Karlsson FA. Early changes in ghrelin following Roux-en-Y gastric bypass: influence of vagal nerve functionality? Obes Surg. 2007;17(3):304–10. 71. Perathoner A, Weiss H, Santner W, Brandacher G, Laimer E, Höller E, Aigner F, Klaus A. Vagal nerve dissection during pouch formation in laparoscopic Roux-Y-gastric bypass for technical simplification: does it matter? Obes Surg. 2009;19(4):412–7. 72. Ponce J, Haynes B, Paynter S, Fromm R, Lindsey B, Shafer A, Manahan E, Sutterfield C. Effect of lap-band-induced weight loss on type 2 diabetes mellitus and hypertension. Obes Surg. 2004;14(10):1335–42. 73. Rubino F, Gagner M, Gentileschi P, Kini S, Fukuyama S, Feng J, Diamond E. The early effect of the Roux-en-Y gastric bypass on hormones involved in body weight regulation and glucose metabolism. Ann Surg. 2004;240(2):236–42. 74. Holst JJ, Madsbad S, Bojsen-Møller KN, Svane MS, Jørgensen NB, Dirksen C, Martinussen C. Mechanisms in bariatric surgery: gut hormones, diabetes resolution, and weight loss. Surg Obes Relat Dis. 2018;14(5):708–14. 75. Stefanidis A, Oldfield BJ. Neuroendocrine mechanisms underlying bariatric surgery: insights from human studies and animal models. J Neuroendocrinol.2017;29(10). https://doi.org/10.1111/ jne.12534. Review. PubMed PMID: 28887853. 76. Stearns AT, Balakrishnan A, Tavakkolizadeh A. Impact of Roux-­ en-­Y gastric bypass surgery on rat intestinal glucose transport. Am J Physiol Gastrointest Liver Physiol. 2009;297(5):G950–7. 77. Nauck MA. Unraveling the science of incretin biology. Eur J Intern Med. 2009;20(Suppl 2):S303–8. 78. Angrisani L, Cutolo PP, Ciciriello MB, Vitolo G, Persico F, Lorenzo M, Scarano P. Laparoscopic adjustable gastric banding with truncal vagotomy versus laparoscopic adjustable gastric banding alone: interim results of a prospective randomized trial. Surg Obes Relat Dis. 2009;5(4):435–8. 79. Burton PR, Brown WA, Laurie C, Hebbard G, O’Brien PE. Criteria for assessing esophageal motility in laparoscopic adjustable gastric band patients: the importance of the lower esophageal contractile segment. Obes Surg. 2010;20(3):316–25. 80. Noel OF, Still CD, Argyropoulos G, Edwards M, Gerhard GS. Bile acids, FXR, and metabolic effects of bariatric surgery. J Obes. 2016;2016:4390254. 81. Tian J, Huang S, Sun S, Ding L, Zhang E, Liu Y, Huang W. Bile acid signaling and bariatric surgery. Liver Res. 2017;1(4):208–13. 82. Evers SS, Sandoval DA, Seeley RJ. The physiology and molecular underpinnings of the effects of bariatric surgery on obesity and diabetes. Annu Rev Physiol. 2017;79:313–34. 83. Tadross JA, le Roux CW. The mechanisms of weight loss after bariatric surgery. Int J Obes. 2009;33(Suppl 1):S28–32. 84. Gerhard GS, Styer AM, Wood GC, Roesch SL, Petrick AT, Gabrielsen J, Strodel WE, Still CD, Argyropoulos G. A role for fibroblast growth factor 19 and bile acids in diabetes remission after Roux-en-Y gastric bypass. Diabetes Care. 2013;36(7):1859–64. 85. Myronovych A, Kirby M, Ryan KK, Zhang W, Jha P, Setchell KD, Dexheimer PJ, Aronow B, Seeley RJ, Kohli R. Vertical sleeve gastrectomy reduces hepatic steatosis while increasing serum bile acids in a weight-loss-independent manner. Obesity (Silver Spring). 2014;22(2):390–400. 86. Nakatani H, Kasama K, Oshiro T, Watanabe M, Hirose H, Itoh H. Serum bile acid along with plasma incretins and serum high-­ molecular weight adiponectin levels are increased after bariatric surgery. Metabolism. 2009;58(10):1400–7. 87. Pournaras DJ, le Roux CW. Are bile acids the new gut hormones? Lessons from weight loss surgery models. Endocrinology. 2013;154(7):2255–6. 5 Physiological Mechanisms of Bariatric Procedures 88. Katsuma S, Hirasawa A, Tsujimoto G. Bile acids promote glucagon-­ like peptide-1 secretion through TGR5 in a murine enteroendocrine cell line STC-1. Biochem Biophys Res Commun. 2005;329(1):386–90. 89. Ryan KK, Tremaroli V, Clemmensen C, Kovatcheva-Datchary P, Myronovych A, Karns R, Wilson-Pérez HE, Sandoval DA, Kohli R, Bäckhed F, Seeley RJ. FXR is a molecular target for the effects of vertical sleeve gastrectomy. Nature. 2014;509(7499):183–8. 90. Després J-P. Excess visceral adipose tissue/ectopic fat the missing link in the obesity paradox? J Am Coll Cardiol. 2011;57(19):1887–9. 91. Madsbad S, Dirksen C, Holst JJ. Mechanisms of changes in glucose metabolism and bodyweight after bariatric surgery. Lancet Diabetes Endocrinol. 2014;2(2):152–64. 92. Wozniak SE, Gee LL, Wachtel MS, Frezza EE. Adipose tissue: the new endocrine organ? A review article. Dig Dis Sci. 2009;54(9):1847–56. 93. Yang R-Z, Lee M-J, Hu H, Pray J, Wu H-B, Hansen BC, Shuldiner AR, Fried SK, McLenithan JC, Gong D-W. Identification of omentin as a novel depot-specific adipokine in human adipose tissue: possible role in modulating insulin action. Am J Physiol Endocrinol Metab. 2006;290(6):E1253–61. 94. de Souza Batista CM, Yang R-Z, Lee M-J, Glynn NM, Yu D-Z, Pray J, Ndubuizu K, Patil S, Schwartz A, Kligman M, Fried SK, Gong D-W, Shuldiner AR, Pollin TI, McLenithan JC. Omentin plasma levels and gene expression are decreased in obesity. Diabetes. 2007;56(6):1655–61. 95. Cammisotto P, Bendayan M. A review on gastric leptin: the exocrine secretion of a gastric hormone. Anat Cell Biol. 2012;45(1):1–16. 96. Fu M, Damcott CM, Sabra M, Pollin TI, Ott SH, Wang J, Garant MJ, O’Connell JR, Mitchell BD, Shuldiner AR. Polymorphism in the calsequestrin 1 (CASQ1) gene on chromosome 1q21 is associated with type 2 diabetes in the old order Amish. Diabetes. 2004;53(12):3292–9. 97. Wiltshire S, Hattersley AT, Hitman GA, Walker M, Levy JC, Sampson M, O’Rahilly S, Frayling TM, Bell JI, Lathrop GM, Bennett A, Dhillon R, Fletcher C, Groves CJ, Jones E, Prestwich P, Simecek N, Rao PV, Wishart M, Bottazzo GF, Foxon R, Howell S, Smedley D, Cardon LR, Menzel S, McCarthy MI. A genomewide scan for loci predisposing to type 2 diabetes in a U.K. population (the diabetes UK Warren 2 repository): analysis of 573 pedigrees provides independent replication of a susceptibility locus on chromosome 1q. Am J Hum Genet. 2001;69(3):553–69. 98. Keim NL, Stern JS, Havel PJ. Relation between circulating leptin concentrations and appetite during a prolonged, moderate energy deficit in women. Am J Clin Nutr. 1998;68(4):794–801. 99. Bai Y, Zhang S, Kim KS, Lee JK, Kim KH. Obese gene expression alters the ability of 30A5 preadipocytes to respond to lipogenic hormones. J Biol Chem. 1996;271(24):13939–42. 100. van Dielen FMH, van’t Veer C, Buurman WA, Greve JWM. Leptin and soluble leptin receptor levels in obese and weight-losing individuals. J Clin Endocrinol Metab. 2002;87(4):1708–16. 101. Faraj M, Havel PJ, Phélis S, Blank D, Sniderman AD, Cianflone K. Plasma acylation-stimulating protein, adiponectin, leptin, and ghrelin before and after weight loss induced by gastric bypass surgery in morbidly obese subjects. J Clin Endocrinol Metab. 2003;88(4):1594–602. 102. Berg AH, Combs TP, Scherer PE. ACRP30/adiponectin: an adipokine regulating glucose and lipid metabolism. Trends Endocrinol Metab. 2002;13(2):84–9. 103. Yang WS, Lee WJ, Funahashi T, Tanaka S, Matsuzawa Y, Chao CL, Chen CL, Tai TY, Chuang LM. Weight reduction increases plasma levels of an adipose-derived anti-inflammatory protein, adiponectin. J Clin Endocrinol Metab. 2001;86(8):3815–9. 75 104. Weyer C, Funahashi T, Tanaka S, Hotta K, Matsuzawa Y, Pratley RE, Tataranni PA. Hypoadiponectinemia in obesity and type 2 diabetes: close association with insulin resistance and hyperinsulinemia. J Clin Endocrinol Metab. 2001;86(5):1930–5. 105. Ouchi N, Kihara S, Arita Y, Maeda K, Kuriyama H, Okamoto Y, Hotta K, Nishida M, Takahashi M, Nakamura T, Yamashita S, Funahashi T, Matsuzawa Y. Novel modulator for endothelial adhesion molecules: adipocyte-derived plasma protein adiponectin. Circulation. 1999;100(25):2473–6. 106. Bäckhed F. Changes in intestinal microflora in obesity: cause or consequence? J Pediatr Gastroenterol Nutr. 2009;48(Suppl 2):S56–7. 107. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The human microbiome project. Nature. 2007;449(7164):804–10. 108. Turnbaugh PJ, Bäckhed F, Fulton L, Gordon JI. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe. 2008;3(4):213–23. 109. Bäckhed F, Manchester JK, Semenkovich CF, Gordon JI. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci U S A. 2007;104(3):979–84. 110. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444(7122):1022–3. 111. Ley RE, Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI. Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A. 2005;102(31):11070–5. 112. Hainer V, Toplak H, Mitrakou A. Treatment modalities of obesity: what fits whom? Diabetes Care. 2008;31(Suppl 2):S269–77. 113. Gagliardi L, Wittert G. Management of obesity in patients with type 2 diabetes mellitus. Curr Diabetes Rev. 2007;3(2):95–101. 114. Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, Burcelin R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes. 2008;57(6):1470–81. 115. Zhang H, DiBaise JK, Zuccolo A, Kudrna D, Braidotti M, Yu Y, Parameswaran P, Crowell MD, Wing R, Rittmann BE, Krajmalnik-Brown R. Human gut microbiota in obesity and after gastric bypass. Proc Natl Acad Sci U S A. 2009;106(7):2365–70. 116. Woodard GA, Encarnacion B, Downey JR, Peraza J, Chong K, Hernandez-Boussard T, Morton JM. Probiotics improve outcomes after Roux-en-Y gastric bypass surgery: a prospective randomized trial. J Gastrointest Surg. 2009;13(7):1198–204. 117. Blanco J, Jiménez A, Casamitjana R, Flores L, Lacy A, Conget I, Vidal J. Relevance of beta-cell function for improved glycemic control after gastric bypass surgery. Surg Obes Relat Dis. 2014;10(1):9–13.. quiz 189–90 118. Dixon JB, Chuang L-M, Chong K, Chen S-C, Lambert GW, Straznicky NE, Lambert EA, Lee W-J. Predicting the glycemic response to gastric bypass surgery in patients with type 2 diabetes. Diabetes Care. 2013;36(1):20–6. 119. Nannipieri M, Mari A, Anselmino M, Baldi S, Barsotti E, Guarino D, Camastra S, Bellini R, Berta RD, Ferrannini E. The role of beta-cell function and insulin sensitivity in the remission of type 2 diabetes after gastric bypass surgery. J Clin Endocrinol Metab. 2011;96(9):E1372–9. 120. Czupryniak L, Wiszniewski M, Szymański D, Pawłowski M, Loba J, Strzelczyk J. Long-term results of gastric bypass surgery in morbidly obese type 1 diabetes patients. Obes Surg. 2010;20(4):506–8. 121. Mari A, Manco M, Guidone C, Nanni G, Castagneto M, Mingrone G, Ferrannini E. Restoration of normal glucose tolerance in severely obese patients after bilio-pancreatic diversion: role of insulin sensitivity and beta cell function. Diabetologia. 2006;49(9):2136–43. 76 122. Camastra S, Gastaldelli A, Mari A, Bonuccelli S, Scartabelli G, Frascerra S, Baldi S, Nannipieri M, Rebelos E, Anselmino M, Muscelli E, Ferrannini E. Early and longer term effects of gastric bypass surgery on tissue-specific insulin sensitivity and beta cell function in morbidly obese patients with and without type 2 diabetes. Diabetologia. 2011;54(8):2093–102. 123. Kashyap SR, Daud S, Kelly KR, Gastaldelli A, Win H, Brethauer S, Kirwan JP, Schauer PR. Acute effects of gastric bypass versus gastric restrictive surgery on beta-cell function and insulinotropic hormones in severely obese patients with type 2 diabetes. Int J Obes. 2010;34(3):462–71. 124. Neel JV. Diabetes mellitus: a “thrifty” genotype rendered detrimental by “progress”? Am J Hum Genet. 1962;14(4):353–62. 125. Gregor MF, Yang L, Fabbrini E, Mohammed BS, Eagon JC, Hotamisligil GS, Klein S. Endoplasmic reticulum stress is reduced in tissues of obese subjects after weight loss. Diabetes. 2009;58(3):693–700. 126. Gregor MG, Hotamisligil GS. Thematic review series: adipocyte biology. Adipocyte stress: the endoplasmic reticulum and metabolic disease. J Lipid Res. 2007;48(9):1905–14. D. R. Funes et al. 127. Algahim MF, Sen S, Taegtmeyer H. Bariatric surgery to unload the stressed heart: a metabolic hypothesis. Am J Physiol Heart Circ Physiol. 2012;302(8):H1539–45. 128. Muskiet MHA, Tonneijck L, Smits MM, van Baar MJB, Kramer MHH, Hoorn EJ, Joles JA, van Raalte DH. GLP-1 and the kidney: from physiology to pharmacology and outcomes in diabetes. Nat Rev Nephrol. 2017;13(10):605–28. 129. Jian Yang MD, Jose PA, Zeng C. Gastrointestinal–renal axis: role in the regulation of blood pressure. J Am Heart Assoc. 2017;6(3):e005536. 130. Docherty NG, le Roux CW. Physiological and pathophysiological signalling between the gut and the kidney: role in diabetic kidney disease. Exp Physiol. 2014;99(9):1138–9. 131. Zhu Z, Xiong S, Liu D. The gastrointestinal tract: an initial organ of metabolic hypertension? Cell Physiol Biochem. 2016;38(5):1681–94. 132. Farah LX, Valentini V, Pessoa TD, Malnic G, McDonough AA, Girardi AC. The physiological role of glucagon-like peptide-1 in the regulation of renal function. Am J Physiol Ren Physiol. 2016;310(2):F123–7. 6 Indications and Contraindications for Bariatric Surgery Christopher DuCoin, Rachel L. Moore, and David A. Provost Chapter Objectives 1. To assess the risk-benefit ratio and indications of bariatric surgery 2. To discuss contraindications to bariatric surgery Introduction Metabolic and bariatric surgery is a proven therapy for the treatment of obesity and obesity-related comorbidities. Available evidence, as detailed elsewhere in the text, strongly suggests that metabolic and bariatric surgery produces weight loss that is significantly greater and more durable than that achieved with the best nonsurgical therapies. This was validated with the 1991 National Institutes of Health (NIH) Consensus Development Conference Statement which set the eligibility criteria for bariatric surgery. To qualify individuals should have a BMI ≥ 40 kg/m2 or a BMI between 35 and 40 kg/m2 if they also have high-risk comorbidities such as severe type 2 diabetes or cardiovascular risk factors [1]. At that time, all of bariatric surgery was being performed via on open laparotomy. Since 1991, bariatric surgery has also been shown to result in resolution or improvement of associated diseases or conditions including, but not limited to, hyperlipidemia, obstructive sleep apnea, polycystic ovarian syndrome, gout, nonalcoholic fatty liver disease, gastroesophageal C. DuCoin (*) Division of Bariatric & General Surgery, University of South Florida, Tampa, FL, USA e-mail: cducoin@health.usf.edu R. L. Moore Moore Metabolics & Tulane University Department of Surgery, New Orleans, LA, USA D. A. Provost Division of General and Bariatric Surgery, Baylor Scott & White Medical Center, Temple, TX, USA reflux, and pseudotumor cerebri. Reductions in the development of cancers, particularly gynecological, breast, and colon cancers, have been demonstrated. Several case-controlled studies have demonstrated improvements in long-term survival. A major benefit of metabolic and bariatric surgery, though not often considered when defining the indications for surgery, is the improvement in overall quality of life. When selecting appropriate candidates for surgery, these benefits must be carefully weighed against the potential perioperative and long-term risks of the procedures. I ndications for Metabolic and Bariatric Surgery Although almost 30 years old, the 1991 National Institutes of Health (NIH) Consensus Development Conference Statement on Gastrointestinal Surgery for Severe Obesity [2] continues to be the most frequently referenced guideline and gold standard when determining the body mass index (BMI) indications for metabolic and bariatric surgery. The statement notes that patients must be acceptable of the operative risks, motivated, well-informed, and able to participate in treatment and follow-up. Patients judged to have a low probability of success with nonsurgical methods of weight loss may be considered for surgery. Assessment of risk-benefit ratio should be performed for each case. Potential candidates include: • Patients whose body mass index (BMI) exceeds 40 kg/m2 • Patients with BMIs between 35 and 40 kg/m2 who also have high-risk comorbid conditions or lifestyle-limiting obesity-induced physical conditions Examples of associated comorbid conditions to be considered in patients with a BMI less than 40 kg/m2 include common conditions such as hypertension, diabetes mellitus, and obstructive sleep apnea to life-threatening cardiopulmonary problems such as obesity-hypoventilation and © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_6 77 78 obesity-­related cardiomyopathy. Physical conditions to be considered include joint disease and gout. The conditions listed were examples and not meant to be all-inclusive. Other obesity-­related comorbidities often considered when determining the appropriateness of surgery in patients with a BMI less than 40 kg/m2 include hyperlipidemia, nonalcoholic fatty liver disease, gastroesophageal reflux, pseudotumor cerebri, asthma, venous stasis disease, and urinary incontinence [3]. Many subspecialty surgeons such as neurosurgeons and orthopedic surgeons will refer their patients for weight loss surgery prior to their definitive procedure. The NIH Consensus Conference Statement noted that surgical candidates should be evaluated by a “multidisciplinary team with access to medical, surgical, psychiatric, and nutritional expertise” [2]. The pros and cons of various treatment options, both surgical and nonsurgical, should be discussed with the patient. Metabolic and bariatric surgery should be performed by a surgeon experienced with the appropriate procedure, working in a program with adequate support for all aspects of perioperative and postoperative care. Postoperative surveillance should continue indefinitely. No upper age limit for bariatric surgery was recommended by the consensus panel. At the time of publication, it was felt that insufficient data was available to make a recommendation for or against surgery in the adolescent population. However, multiple studies have now shown bariatric surgery to be safe in both extremes of age. Defining the indications for metabolic and bariatric surgery begins with an assessment of the risk-benefit ratio of a given procedure. Significant advances in surgical techniques, reductions in operative risk, and greater knowledge of the potential risk of untreated obesity have greatly altered the risk-benefit ratio of surgery since 1991. Many of these changes were summarized in the 2004 American Society for Bariatric Surgery (ASBS) Consensus Conference Statement on Bariatric Surgery for Morbid Obesity [4] and include: 1. The marked increase in the incidence of obesity 2. Expansion of available operative procedures (e.g., vertical sleeve gastrectomy and laparoscopic adjustable gastric banding) 3. Significant reductions in perioperative mortality and morbidity 4. The introduction of laparoscopic techniques 5. Increased experience with a team management approach 6. Increased experience with metabolic and bariatric surgery in adolescents and the elderly 7. Greater demonstration of the effect of surgery in improving or reversing obesity-related comorbidities 8. Demonstration that metabolic and bariatric surgery improves life expectancy The 1991 NIH guidelines currently preclude individuals with a BMI between 35 and 40 kg/m2 without comorbidities C. DuCoin et al. or with a BMI < 35 kg/m2 from bariatric surgery. With the development of “accredited” bariatric programs and surgeons with focused bariatric training, there has been a large reduction in the morbidity and mortality associated with these procedures. The health and mortality risk of untreated obesity is better defined and greater than was appreciated in 1991. The result is a shift in the risk-benefit ratio, again, favoring surgery and leading to the consideration of lowering the BMI indications for metabolic and bariatric surgery. The American Society for Metabolic and Bariatric Surgery (ASMBS), the Obesity Society, the American Association of Clinical Endocrinologists (AACE) [2], the International Diabetes Federation (IDF), and the American Diabetes Association (ADA) [5] have endorsed guidelines recommending consideration of metabolic and bariatric surgery in patients with a BMI of 30–34.9 kg/m2 with diabetes or metabolic syndrome. The IDF position statement from March 2011 recommends that surgery should be considered as an alternative treatment option in patients with a BMI between 30 and 35 kg/m2 when the diabetes cannot be adequately controlled with an optimal medical regimen, especially in the presence of other major cardiovascular disease risk factors [5]. Multiple studies also support that BMI alone is not the perfect predictor for severity of obesity. Recently waist circumference has been used as both a surrogate and in conjunction with BMI. Values used for waist circumference include 40 in. (102 cm) for men and 35 in. (88 cm) for women. When BMI and waist circumference are used together, it has been found that those with more central obesity have a higher risk of disease and death. Recently, a variety of weight loss devices have been introduced to the market, each with their own criteria for use. In 2001 the US Food and Drug Administration (FDA) approved the Lap-Band™ laparoscopic adjustable gastric band for patients with a BMI between 30 and 34.9 kg/m2 and an obesity-­ related comorbidity. In 2015 the FDA approved three intragastric balloon systems for patients with a BMI ranging between 30 and 40 kg/m2. Although the FDA took steps to lower the BMI criteria for these implantable devices, the organization did not do so for all implantable weight loss devices. The vagal nerve blocking therapy (VBLOC) was approved in 2015, and the BMI requirements were identical to the 1991 NIH guidelines. Approved in 2016, the AspireAssist system, a gastric-emptying device, was approved for a BMI range between 35 and 55 kg/m2. ontraindications to Metabolic and Bariatric C Surgery While there are few absolute contraindications to bariatric and metabolic surgery, there are no absolute contraindications specific to bariatric surgery [6]. Most would be included 6 Indications and Contraindications for Bariatric Surgery in lists of contraindications to any elective surgical procedure. Relative contraindications to surgery may include severe heart failure, unstable coronary artery disease, end-­ stage lung disease, active cancer diagnosis or treatment, cirrhosis with portal hypertension, uncontrolled drug or alcohol dependency, and severely impaired intellectual capacity [6]. Crohn’s disease may be a relative contraindication to RYGB [7] and BPD [8]. Poor results following a variety of bariatric surgical procedures have been reported in patients with lower levels of mental capacity. This inability to comprehend the treatment process is considered a contraindication by most surgeons. Patients deemed a prohibitive operative risk should not be offered surgery including those with contraindications to general anesthesia or uncorrectable coagulopathy. Bariatric surgery should not be performed on patients with limited life expectancy. Metabolic and bariatric surgery should be postponed in patients with active peptic ulcer disease until successful treatment has been confirmed. Patients who are pregnant or who expect to be pregnant within 12–18 months of surgery should be deferred. Patients who are on the transplant list or who have received transplants are candidates for bariatric surgery [9]. In addition, patients must be able, willing, and motivated to comply with postoperative lifestyle changes, dietary supplementation, and follow-up. Specific Considerations Extremes of Age The 1991 NIH Consensus Conference Statement did not include an upper age limit for surgery for obesity although many programs and authors have restricted metabolic and bariatric surgery to patients less than 65 years of age. While a few publications have noted increasing age to be a risk factor for postoperative complications, both ASMBS & SAGES, along with numerous publications, have demonstrated that surgery can be performed safely and effectively in the older patient population [10, 11]. A review of perioperative outcomes following bariatric surgery in nearly 50,000 patients in the American College of Surgeons (ACS) National Surgical Quality Improvement Program database demonstrated no significant increase in mortality in elderly patients [10]. While improvements in longevity resulting from surgery are difficult to demonstrate, improvements in obesity-­ related comorbidities and quality of life justify performing metabolic and bariatric surgery in appropriately selected patients aged 65 and older. Sufficient evidence has accumulated to consider surgery for severe obesity an appropriate therapeutic option in properly selected adolescents [12]. Selection criteria and specific 79 considerations in the preoperative evaluation and postoperative management are addressed in greater detail in Chap. 7. As the effects on maturation and growth are unknown, metabolic and bariatric surgery in the preadolescent is considered experimental and is not recommended. Psychiatric Illness Patients referred for bariatric surgery have a higher incidence of obsessive-compulsive disorder, substance abuse/dependency, binge eating disorder, post-traumatic stress disorder, generalized anxiety disorder, depression, and schizophrenia when compared to the general population [13]. Patients with poorly managed psychiatric disorders may have a suboptimal outcome after bariatric surgery [13]. However, no consensus recommendations exist regarding preoperative psychological evaluation [14, 15]. A diagnosis of psychopathology, including eating disorders, does not preclude metabolic and bariatric surgery [16]. Successful outcomes have been demonstrated in patients with major depressive disorder, bipolar disorder, stable schizophrenia, and binge eating. A preoperative psychological evaluation in patients with a history of psychiatric illness or a positive screening is beneficial in assessing the patient’s emotional stability at the time of surgery and ensuring adequate support during the preoperative and postoperative phases of surgery [17]. Patients with active psychosis or recent hospitalization for psychosis, as well as patients with suicidal ideation or recent suicidal attempts, should have surgery delayed or postponed and treatment initiated. However, this does not preclude a patient from becoming a surgical candidate once the disease process is stable. Ongoing therapy for these patients is essential in the postoperative period if deemed appropriate for surgery. Cirrhosis Nonalcoholic fatty liver disease is common in severe obesity, with histologic evidence of steatosis present in nearly 90% of patients undergoing metabolic and bariatric surgery and unexpected cirrhosis identified in 2% of patients [18]. Weight loss following surgery has been demonstrated to improve the histologic findings of steatosis and steatohepatitis. Bariatric surgery has also shown potential to halt and even reverse liver damage, while liver functions largely remained stable [19]. Surgery may be safely performed in patients with stable cirrhosis [20]. When cirrhosis is an incidental finding at surgery, it is recommended to proceed in the absence of findings of significant portal hypertension including severe ascites and perigastric varices. If evidence of portal hypertension is encountered unexpectedly, the procedure should be aborted. Essentially, patients with decompensated cirrhosis are far 80 C. DuCoin et al. more likely to suffer adverse outcome than those with compensated disease after bariatric surgery [21]. Bariatric surgery has been reported in highly selected patients with advanced cirrhosis in preparation for liver transplantation. Reported cases are few and should only be performed in tertiary centers in partnership with a liver transplant service. Question Section Nonambulators References Nonambulatory status is considered a relative contraindication to bariatric surgery by some programs. Although nonambulatory status and poor functional capacity have been demonstrated to increase perioperative morbidity and reduce postoperative weight loss, this increased risk does not exceed the potential benefit in properly selected, motivated patients. 1. Gastrointestinal surgery for severe obesity: National Institutes of Health Consensus Development Conference Statement. Am J Clin Nutr. 1992; 55(2 Suppl):615S–9S. 2. NIH conference. Gastrointestinal surgery for severe obesity. Consensus development conference panel. Ann Intern Med. 1991;115:956–61. 3. Mechanick JI, Youdim A, Jones DB, Garvey WT, Hurley DL, McMahon MM, et al. Clinical practice guidelines for the perioperative nutritional, metabolic, and nonsurgical support of the bariatric surgery patient – 2013 update: cosponsored by American Association of Clinical Endocrinologists, The Obesity Society, and American Society for Metabolic & Bariatric Surgery. Surg Obes Relat Dis. 2013;9:159–91. 4. Buchwald H. 2004 ASBS consensus conference: consensus conference statement – bariatric surgery for morbid obesity: health implications for patients, health professionals, and third-party payors. Surg Obes Relat Dis. 2005;1:371–81. 5. Dixon JB, Zimmet P, Alberti KG, Rubino F. Bariatric surgery: an IDF statement for type 2 diabetes. Surg Obes Relat Dis. 2011;7:433–47. 1. The following should all be considered contraindications for bariatric surgery except: A. Inability to undergo general anesthesia B. Limited life expectancy due to irreversible cardiopulmonary disease or inoperable malignancy C. Inability to comprehend the risks and benefits of the planned procedure and comply with postoperative HIV Infection lifestyle and dietary modifications and follow-up D. HIV positivity Patients with human immunodeficiency virus (HIV) infec- 2. Which of the following psychiatric conditions is a contration were once considered inappropriate candidates for metindication for bariatric surgery? abolic and bariatric surgery due to the expected progression A. Bipolar disorder to acquired immunodeficiency syndrome (AIDS) and AIDS-­ B. Active psychosis or recent suicidal ideation associated cachexia. Modern antiretroviral therapies have C. Stable schizophrenia dramatically reduced disease progression, extending life D. Binge eating disorder expectancy, often with nearly undetectable viral loads. 3. Which age, comorbid condition, and BMI is a contraindiAntiretroviral-induced lipodystrophy has contributed to the cation to surgery? incidence of obesity in the HIV population, with 20% of A. 56-year-old with steatohepatitis and a BMI of 41 kg/ patients progressing from normal/overweight to obesity m2 within 2 years of starting antiretroviral therapy. B. 67-year-old with HTN and a BMI of 36 kg/m2 Reported case series have demonstrated the safety and C. 47-year-old with perigastric varices and a BMI of efficacy of metabolic and bariatric surgery in patients with 67 kg/m2 well-controlled HIV infection [22, 23]. Patients with HIV D. 19-year-old with OSA and a BMI of 41 kg/m2 infection considered for surgery should demonstrate a stable, 4. Which procedure is approved for a BMI < 35 kg/m2? appropriate response to antiretroviral therapy as determined A. Sleeve gastrectomy by CD4 counts. Preoperative and postoperative consultation B. Intragastric balloon with an HIV infectious disease specialist is mandatory. HIV C. Roux-en-Y gastric bypass and AIDS is not a contraindication to surgery. D. AspireAssist Conclusion Metabolic and bariatric surgery produces durable weight loss well beyond that achieved with medical and behavioral therapies, with resultant improvement in obesity-related comorbidities and quality of life. Advances in the field have led to an expansion of the indications for surgery. Appropriate patient selection is mandatory to ensure optimal results while minimizing perioperative risks. 6 Indications and Contraindications for Bariatric Surgery 6. SAGES Guidelines Committee endorsed by ASMBS, Guideline for the Clinical Application of Laparoscopic Bariatric Surgery, 25 March 2008. https://www.sages.org/publications/guidelines/guidelines-for-clinical-application-of-laparoscopic-bariatric-surgery/. 7. Pretolesi F, Camerini G, Marinari GM, et al. Crohn disease obstruction of the biliopancreatic limb in a patient operated for biliopancreatic diversion for morbid obesity. Emerg Radiol. 2006;12(3):116–8. 8. Parikh M, Duncombe J, Fielding GA. Laparoscopic adjustable gastric banding for patients with body mass index of ≤ 35 kg/m2. Surg Obes Relat Dis. 2006;2(5):518–22. 9. Gheith O, Al-Otaibi T, Halim MA, et al. Bariatric surgery in renal transplant patients. Exp Clin Transplant. 2017;15(Suppl 1):164–9. 10. Dorman RB, Abraham AA, Al-Refaie WB. Bariatric surgery outcomes in the elderly: an ACS NSQIP study. J Gastrointest Surg. 2012;16:35–44. 11. Lainas P, Dammaro C, Gaillard M, et al. Safety and shortterm outcomes of laparoscopic sleeve gastrectomy for patients over 65 years old with severe obesity. Surg Obes Relat Dis. 2018. pii: S1550-­ 7289(18)30122-9; https://doi.org/10.1016/j. soard.2018.03.002.. [Epub ahead of print]. 12. Michalsky M, Reichard K, Inge T, Pratt J, Lenders C. Update: ASMBS pediatric committee best practice guidelines. Surg Obes Relat Dis. 2012;8:1–7. 13. Kinzl JF, Schrattenecker M, Traweger C, et al. Psychosocial predictors of weight loss after bariatric surgery. Obes Surg. 2006;16(12):1609–14. 14. Bauchowitz AU, Gonder-Frederick LA, Olbrisch ME, et al. Psychosocial evaluation of bariatric surgery candidates: a survey of present practices. Psychosom Med. 2005;67:825–32. 81 15. Van Hout GC, Verschure SK, van Heck GL. Psychosocial predictors of success following bariatric surgery. Obes Surg. 2005;15:552–60. 16. LeMont D, Moorehead MK, Parish MS, Reto CS, Ritz SJ. Suggestions for the pre-surgical psychological assessment of bariatric surgery candidates. ASMBS 2004.; http://asmbs. org/2012/06/pre-surgical-psychological-assessment/. 17. Pull CB. Current psychological assessment practices in obesity surgery programs: what to assess and why. Curr Opin Psychiatry. 2010;23:30–6. 18. Marceau P, Biron S, Hould FS, Marceau S, Simard S, Thung SN, Kral JG. Liver pathology and the metabolic syndrome X in severe obesity. J Clin Endocrinol Metab. 1999;84:1513–7. 19. Jan A, Narwaria M, Mahawa K. A systematic review of bariatric surgery in patients with liver cirrhosis. Obes Surg. 2015;25(8):1518–26. 20. Shimizu H, Phuong V, Maia M, Kroh M, Chand B, Schauer PR, et al. Bariatric surgery in patients with liver cirrhosis. Surg Obes Relat Dis. 2013;9:1–6. 21. Mosko JD, Nguyen GC. Increased perioperative mortality following bariatric surgery among patients with cirrhosis. Clin Gastroenterol Hepatol. 2011;9(10):897–901. 22. Flancbaum L. Initial experience with bariatric surgery in asymptomatic human immunodeficiency virus-infected patients. Surg Obes Relat Dis. 2005;1:73–6. 23. Selke S, Norris S, Osterholzer D, Fife KH, DeRose B, Gupta SK. Bariatric surgery outcomes in HIV-infected subjects: a case series. AIDS Patient Care STDs. 2010;24:545–50. 7 Preoperative Care of the Bariatric Patient Renée M. Tholey and David S. Tichansky Chapter Objectives 1. Describe evidence-based preoperative evaluation of the bariatric patient. 2. Discuss risk assessment to optimize patient selection. 3. Discuss informed consent. 4. Explain the establishment of preoperative care pathways. Introduction Preoperative care of the bariatric patient starts before the patient arrives. Establishment of data-driven patient selection protocols and preoperative evaluation pathways not only streamlines practice but also improves patient safety. Both evaluation and individualized risk assessment are essential for achieving best outcomes and allowing the patient to give a truly informed consent. Ideally, comprehensive informed consent would include specific outcome assessments based on the patient’s own metabolic acuity and known outcomes for weight loss and comorbidity resolution. Best preoperative care will yield a comprehensive understanding of a patient’s medical history as it pertains to predicted outcomes, cardiac health, venous thromboembolism risk, sleep architecture and pulmonary function, gastroesophageal anatomy and Helicobacter pylori status, and psychological ability to comply with required postoperative recommendations. Regardless of whether a specific evaluation in question is subjective or objective, it should be standardized in an evidence-­based protocol. This chapter will R. M. Tholey (*) ∙ D. S. Tichansky Department of Surgery, Thomas Jefferson University Hospital, Philadelphia, PA, USA e-mail: Renee.Tholey@jefferson.edu describe evidence-­based comprehensive preoperative evaluation of the bariatric patient, discuss risk assessment to optimize patient selection and informed consent, and explain establishment of preoperative pathways. Patient Selection Perhaps the most important part of the preoperative evaluation is patient selection. Ideally, patient selection is a dynamic process, rather than a single point-in-time decision. During the initial consultation, the surgeon and care team should take a thorough history and physical and determine if there are any immediate contraindications to surgery. If the patient is acceptable at that point, it only means they are acceptable to continue the preoperative evaluation. This workup should be conservative and data informed. Ultimately, the increase in the acceptance of weight loss surgery by the medical community and the public is likely based on improved outcomes. These outcomes are partially based on improved understanding of the risk and benefits of weight loss surgery. Bariatric surgery patient selection is still based on the National Institute of Health Consensus Statement on Gastrointestinal Surgery for Severe Obesity. This states that patients with a body mass index (BMI) of 40 kg/m2 or greater, or patients with a BMI of 35 kg/m2 or greater with an obesity-related comorbidity, will likely benefit from weight loss surgery [1]. Please keep in mind this consensus was reflective of a time when open surgery was prevalent and different bariatric procedures were being performed, versus the more commonly done sleeve gastrectomy today. However, the consensus provides a good starting point, and other organizations have elected to follow these guidelines. For example, the International Diabetes Federation Taskforce in 2011 concordantly recommended bariatric surgery be considered for adults with a BMI ≥ 35 and Type 2 diabetes [2]. BMI cutoffs aid in selection criteria and have been largely adopted by insurance companies, but what about the postoperative risks as BMI increases? Multiple studies have documented © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_7 83 84 that as the BMI increases so do the postoperative complications, especially as those BMIs became more extreme, for example, greater than 70 kg/m2 [3]. While it seems nonintuitive to consider extreme weight an exclusion criterion for weight loss surgery, it should raise concern that these patients are not average-risk and require more extensive preoperative evaluation. Several studies have also shown that male gender and hypertension have been associated with increased ­mortality [4]. There are many more data points to examine beyond BMI, gender, and hypertension. For example, the patient’s age and mobility should be taken into account. Finks et al. demonstrated that age greater than 50 and mobility limitations both independently increased the risk of a p­ ostoperative complication in a large study of 2075 patients undergoing gastric bypass [5]. A more recent study used the Metabolic and Bariatric Surgery Accreditation and Quality Improvement Program (MBSAQIP) data set and again found that immobile patients had a greater risk of mortality and multiple complications regardless of procedure type, including both the gastric bypass and the sleeve gastrectomy [6]. Conversely, another study did not find any difference in perioperative mortality in immobile patients undergoing bariatric surgery but did find statistically significant less improvement in diabetes, hypertension, and obstructive sleep apnea in the wheelchair bound group [7]. The question of age and mobility limitations can be determined at the programmatic level. Should there be an age cutoff in your practice? Should you refuse to even consider surgery on a patient who cannot ambulate? The literature appears to mostly support an affirmative answer should you choose that path or at least an informed discussion for the patient regarding these issues should you choose not to enforce cutoffs. There are other issues that predispose the patient to increased risk. For example, venous thromboembolism (VTE) risk must be evaluated, as well as cardiac risk and obstructive sleep apnea. These disease processes and how they affect the bariatric patient are discussed in depth in the next few sections. Cardiac Evaluation Evaluation of cardiac status and cardiac risk preoperatively is one of the essential elements in promoting safety in any surgical patient, but especially in morbidly obese patients. Obesity is associated with multiple comorbidities including diabetes, obstructive sleep apnea, dyslipidemia, and hypertension. All of these conditions can potentially contribute to severe cardiovascular events such as heart failure, arrhythmias, and sudden cardiac death. Excessive adipose accumulation increases cardiac workload by increasing sympathetic tone and heart rate as well as filling pressure leading to cardiac failure [8]. R. M. Tholey and D. S. Tichansky There is evidence that obesity has a paradoxical (protective) effect on mortality in patients presenting with STEMI, as a normal BMI is actually a predictor of inducible-VT [9]; however, increased BMI is also shown to predict increased cardiovascular mortality sometimes two to four times higher than normal weight individuals [10]. Rates of cardiac arrest and annualized mortality were found to be substantially higher in those undergoing bariatric surgery when compared with other forms of general surgery [11]. Preoperative workup should include history of coronary artery disease, coronary symptoms, and risk factors (diabetes, smoking, hypertension, etc.). Anyone over the age of 50 certainly requires cardiac evaluation. A physical and electrocardiogram (ECG) are then performed. If the patient’s age is less than 50 with a normal ECG and no cardiac risk factors other than obesity, they fall into the low-risk stratification for a perioperative cardiac event. Regarding ECG interpretation, bariatric patients are more likely to have an increased QT interval which could degenerate into torsade de pointes [12]. This is important to recognize as several drugs used perioperatively in gastrointestinal surgery can produce prolonged QT. Patients who have a history of heart failure, cerebrovascular disease, renal insufficiency, diabetes, or compensated ischemic heart disease will likely require stress testing to accurately evaluate left ventricular function according to the American College of Cardiology (ACC) and American Heart Association (AHA) Guidelines. Should the patient have had a recent intervention requiring a drug-eluting stent, it would be prudent to wait 6 months before attempting an elective procedure to decrease the risk of stent thrombosis and a cardiac event [13]. Aspirin should be continued perioperatively [14]. If the patient is on antiplatelet therapy, then the ACC/ AHA 2014 guidelines recommend that the management of discontinuing antiplatelet therapies is at the discretion of the surgeon and cardiologist and to continue aspirin if possible. We recommend that cardiac risk assessment be left to the expertise of a board-certified cardiologist. Bariatric surgeons should collaborate with cardiologists using evidence-based risk stratification. Test results should be available to anesthesia and the cardiology team chosen should be available postoperatively should the need arise. Preoperative VTE Evaluation The second leading cause of death after leaks in bariatric surgery patients is pulmonary embolism (PE) and is responsible for 40–50% of fatalities. Venous thromboembolism (VTE) rates, which include both PE and deep venous thrombosis (DVT), are reported to be between 0.25% and 3.5%. The majority of patients, approximately 97%, have a predicted risk of less than 1% of developing VTE, but identifying the other 2.5% of patients is critical. Among 45 examined 7 Preoperative Care of the Bariatric Patient variables, a final risk-assessment model contained 10 variables that were predictive of post-discharge VTE including congestive heart failure, paraplegia, reoperation, dyspnea at rest, nongastric band surgery, age ≥60, male sex, BMI ≥50 kg/m2, hospital stay ≥3 days, and operative time ≥3 h [15]. This study utilized a risk calculator and suggested that a calculated 0.4% risk should translate to 2 weeks of postdischarge prophylaxis, and this incorporated approximately 20% of patients. Very high risk was considered to be a calculated risk of >1% and would mandate 4 weeks of anticoagulation. The risk calculator can be found here http:// www.r-calc.com under the formulas tab. Another scoring system, the Michigan Bariatric Surgery Collaborative, divides patients into low (<1%), moderate (1–4%), and high risk (>4%) and provides guidelines for chemical thromboprophylaxis based on this stratification [16]. Another recent study found that the major risk factors for VTE included a prior history of VTE, heart failure, postoperative complications, and open surgery with VTE rates at 30 and 90 days equaling 0.34% and 0.51% [17]. They found that no use of postoperative chemoprophylaxis was an independent predictive factor of VTE. None of these studies provide specific dosing strategies. An even more rare concern is porto-mesenteric venous thrombosis with median development being 14 days post-op [18]. The most common cause of coagulopathy disorders was protein C and/or S deficiency followed by prothrombin gene mutation. It is therefore important to remember to take a thorough history. Any patient with a past history of VTE or a personal family history of the relatively uncommon hypercoagulable states such as factor V Leiden, protein C and S deficiency, and protein C resistance also increases the risk for VTE. Lastly the use of inferior vena cava filters for prevention has largely fallen out of favor at most institutions. Several studies have demonstrated that there is no evidence to suggest that the benefits of the filter outweigh the risks [19, 20]. In addition to anticoagulants, the risk of VTE can be decreased further by smoking cessation, increasing ambulation, and use of thromboembolism-deterrent (TED) hose and sequential compression devices perioperatively. Anticoagulation can be proved to all patients postoperatively for a duration of at least 14, if not 30, days or can be more specifically individualized to each patient using the risk calculators cited above. leep Apnea and Obesity Hypoventilation S Evaluation The incidence of obstructive sleep apnea (OSA) in the morbidly obese population is quoted at anywhere from 71% to 95% depending on the patient’s BMI [21]. This places bariatric 85 patients at significant risk for ischemic heart disease, hypertension, cerebrovascular accidents, and cardiac arrhythmias. Of particular interest to surgeons is the potential for postoperative respiratory issues including hypoxia, hypercapnia, and bronchospasm and the need for reintubation. This risk is exacerbated by the use of anesthetics and narcotic medications in the postoperative period. Hypercapnia as a component of obesity hypoventilation syndrome (OHS) puts patients with this syndrome at particular risk for becoming hyper-somnolent due to carbon dioxide narcosis. The gold standard for evaluation is nocturnal polysomnography (PSG). During PSG, the number of apneic episodes can be quantitated. The apnea-hypopnea index (AHI) indicates the abscess of sleep apnea if less than 5, mild OSA for 5–15, moderate OSA for >15, and severe OSA if >30. However, ordering PSG routinely for every bariatric patient is not cost-effective or appropriate. A consensus panel was formed to specifically address this issue. The panel provided 58 statements as recommendations for the guideline [22]. They determined that the Epworth Sleepiness Score (ESS) should not be used as a screening tool. They instead proposed that the STOP-Bang score be used to stratify high-risk OSA [23]. The STOP-Bang score includes risk factors for OSA such as snoring, daytime sleepiness, observed apnea, hypertension, age >50, neck circumference, and male gender. A score ≥3 indicates a moderate risk of OSA. The panel also recommended venous HCO3 be used as part of the routine screening tool for coexistence of OHS and that OHS should be screened for in bariatric patients with OSA (coexistence 20%). Patients with neuromuscular disorders or obstructive lung disease should be considered as this may increase perioperative hypoventilation risk. Lastly, they stated that the oxygen desaturation index, which is the number of times per hour of sleep that the blood oxygen level drops below baseline, is reliable for detection of OSA. If sleep apnea is diagnosed, a titration study is indicated to determine the appropriate setting of a continuous positive airway pressure (CPAP) device that can help maintain an open airway when the patient is asleep. Patients should be encouraged to bring this mask with them for use in the hospital after their surgery. Another set of guidelines from May 2012 can be found on the American Society for Metabolic and Bariatric Surgery (ASMBS) website. Obesity hypoventilation syndrome (OHS) as mentioned above is present in a subset of patients with OSA. The diagnosis is made in obese patients with a pCO2 >45 in the absence of other respiratory or neuromuscular disorders. A serum bicarbonate level of >27 is sensitive but not specific for elevated carbon dioxide. These patients suffer more profound hypoxemia when asleep. Selected patients such as those on home oxygen or those who have severe COPD may need formal pulmonary function testing prior to surgery. 86 Patients with severe pulmonary dysfunction are at risk for prolonged mechanical ventilation, tracheostomy, and higher mortality. A good understanding of the patient’s preoperative pulmonary function is necessary to adequately assess this subset of patients for surgery. valuation of Upper Gastrointestinal E Anatomy The decision on whether to evaluate patients preoperatively with an esophagogastroduodenoscopy (EGD) or an upper gastrointestinal series (UGI) is still controversial and varies between bariatric centers. Certainly patients undergoing revision surgery will need both an UGI and EGD to define the anatomy and evaluate for reflux or ulcers. If there is severe reflux or dysmotility suspected, then, rarely, manometry and pH studies may be needed. The controversy comes in for patients undergoing a primary bariatric procedure. Some studies suggest a preoperative EGD in all patients. An analysis of 801 patients found abnormal EGD findings in 65% of patients, most commonly gastritis (32%) and gastroesophageal reflux (24%) [24]. Malignancies were observed in 0.5% of patients. Helicobacter pylori were diagnosed in 3.7%. Other studies suggest EGD be performed only in those patients with symptoms as the EGD is more likely to yield pathology [25]. Conversely, some studies recommend against EGD especially in patients undergoing a sleeve gastrectomy. One such study found that 89% of scopes were completely normal, and no cancers were found [26]. Another study evaluated the use of a preoperative swallow and recommends against it [27]. The researchers found that the preoperative swallow does not offer any advantage over selective intraoperative hiatal exploration in the discovery of a hiatal hernia. In fact they found that when there is a false positive result the surgery is slightly prolonged. A meta-analysis identified 532 citations and included 48 studies to reveal that the proportion of EGDs resulting in a change in surgical management was 7.8% [28]. After removing benign findings with a controversial impact on management (hiatal hernia, gastritis, peptic ulcer), this was found to be 0.4%. Changes in medical management were seen in 27.5%, but after eliminating Helicobacter pylori infection, these were found to be 2.5%. Based on the aforementioned data the following recommendations are reasonable. For symptomatic patients, EGD is recommended. EGD is preferable to UGI and can in addition allow a biopsy for Helicobacter pylori. For asymptomatic patients undergoing a sleeve gastrectomy, an EGD is not required. For operations in which the intestinal anatomy is excluded, such as a RYGB or biliopancreatic diversion with R. M. Tholey and D. S. Tichansky duodenal switch, an EGD is recommended. All patients undergoing revision should have evaluation of the anatomy using EGD, UGI, or both. Regarding Helicobacter pylori infection, an EGD is not warranted solely for this reason as breath tests or stool antigens are available. Routine screening for Helicobacter pylori is recommended for high-prevalence areas. When performing an EGD, it is important to keep in mind that undiagnosed OSA may be observed when the patient undergoes conscious sedation. Psychological Evaluation All patients should undergo a preoperative psychological evaluation prior to bariatric surgery. This accomplishes several things. First, it screens for the few patients for whom surgery is contraindicated whether due to severe psychiatric disease such as schizophrenia or due to an inability to understand and adhere to the postoperative instructions. Second, it identifies those patients that may have a temporary contraindication such as undertreated depression, psychosis, or bipolar disorder. These patients may require several weeks to months of therapy to eventually proceed to surgery and reach the best postoperative outcome. Psychiatric screening also evaluates for those patients that meet criteria for food addiction. One study found that 16.9% of patients met criteria for food addiction and 15–40% endorsed emotional eating [29]. Emotional eating may affect outcomes although further research needs to be done in this area. A second study found food addiction is present in 25% of patients and that these patients had a significantly higher prevalence of mood and anxiety disorders as well as suicidality [30]. The study did not find a link between food addiction and current alcohol addiction. Of note psychological screening also delves into present or past alcohol or drug abuse. Bariatric patients are at high risk of “addiction transfer” substituting food for a new addiction such as alcohol or drugs following surgery [31]. Often patients want to use their own psychologist, and this should be discouraged for two reasons. First, many therapists do not know how to perform a comprehensive bariatric evaluation and may not be familiar with how to counsel our particular patient population postoperatively. Second, if there is a failure of therapy, therapists may be reluctant to highlight that by rejecting someone for surgery. We therefore recommend that a psychologist that can perform a high-quality pre-bariatric assessment be mandated and the resulting assessment be communicated with the surgical team in a multidisciplinary fashion or via a medical letter that summarizes the main conclusions of the evaluation. 7 Preoperative Care of the Bariatric Patient Informed Consent Truly informed consent requires a consent that is individualized to each patient. Upon completion of the preoperative workup, the benefit-risk ratio that was discussed at the patient’s initial evaluation may have changed. If so, these findings must be discussed with the patient to obtain fully informed consent. Of note, Madan et al. showed that patients often forget significant elements of their preoperative teaching and education including the most common postoperative complications [32]. Thus, the consent process should ideally be another full discussion of the potential risks, including any extra risks those individualized to the patient, at their preoperative visit. It is a physician’s legal obligation that the patient has all of the information required to make an informed decision. Many institutions, and some states through legislation, are now requiring the physician, rather than another licensed healthcare provider such as a nurse practitioner, obtain surgical consent from the patient. Physicians should present their outcomes in the context of the national outcomes as well as risks and alternatives being offered to the patient. The provider obtaining consent should be familiar with the content of the forms and should review the forms with the patient. An individualized form specific to the procedure is preferred over the institution’s generic consent form. The language in the consent form should be in simple terms. As lack of patient memory or understanding of the content of a consent form can sometimes discredit the multiple releases theoretically provided on signed standardized consent forms, it is essential the patient convey an understanding of what they are signing. The patient should be given a last chance to ask any and all questions prior to consenting. The surgeon should perform documentation of the specifics of this discussion. The ultimate consent privilege lies with the patient alone. Once consent is obtained, the patient still retains the legal and ethical right to revoke the consent as well as ask additional questions to reinforce it. It is prudent for patients to be educated and express understanding for truly informed consent. Conclusion: Standardized Care Pathways In summary, it is essential to set up practice standards such that each patient can be approached in a standardized, evidence-­ based fashion preoperatively to optimize care. Recent literature reviews imply a decrease in hospital complications associated with care pathways. In one recent comparison of 65 patients who were treated in a care pathway versus 64 patients who were not, the pathway group had Foley catheters removed earlier, were mobilized on the surgery day more often, used spirometers more often, and 87 had nutrition conducted in a better fashion [33]. While clinical pathways are typically used in the in-hospital setting, they can also enhance care in the preoperative setting. It is likely the surgeon’s data review involved in setting up these pathways is at least partially what improves care by better informing the surgeon’s practice. Also a comprehensive data review followed by standardized preoperative protocols will limit errors of omission and inappropriate offerings of surgery, ensuring no patient gets left behind or falls through the cracks while increasing efficiency and avoiding unnecessary testing. In this manner, risk of process failure is minimized and patient risk is reduced resulting in improved quality of care. Hence, this chapter recommends that the surgeon who has not yet developed a formal preoperative pathway should perform a focused examination of their practice patterns, processes, and outcomes. These elements should then be compared to what is data driven in the literature. Discrepancies between the two should be corrected by changes in practice or justified objectively by variations in local standards of care. Once done, the final resolutions should be written down and followed as a formal standardized protocol for patient selection, a multisystem preoperative evaluation, and informed consent. Gaining acceptance by staff and collaborating physicians is often straightforward when offered in the context of education. Lastly, these standards have to be “living” documents and periodically reviewed to ensure that they evolve as new data emerges. As these pathways were recommended in previous “excellence” initiatives, they will likely be present in the new era of quality assessment and improvement. Question Section 1. All of the following patient characteristics have been shown to increase the risk of perioperative complications except: A. Male gender B. Age >50 C. Hypertension D. Black race E. Inability to walk 200 ft 2. Most current data support the following except: A. Prophylactic IVC filter in someone with a history of DVT B. Cardiac stress test in someone with a history of heart failure C. Preoperative and postoperative use of CPAP in a patient with an AHI >15 D. Not offering surgery to someone with undertreated bipolar disease E. Obtaining a sleep study with a STOP-Bang score ≥3 88 3. A preoperative EGD is recommended for the following patients: A. Patients undergoing revision surgery B. Patients with a history of reflux C. Patients undergoing procedures that exclude the stomach D. Patients with a history of ulcer disease E. All of the above 4. Truly informed consent involves all of the following except: A. Explanation of risks and benefits B. Discussion of the patient’s individualized risk profile C. Demonstrated fifth-grade reading level by the patient D. Expressed understanding by the patient E. The consent form written in simple terms References 1. Gastrointestinal Surgery for Severe Obesity. NIH Consensus Statement 1991. Accessed July 2018; Available from: http://consensus.nih.gov/1991/1991GISurgeryObesity084html.htm. 2. Dixon JB, et al. Bariatric surgery: an IDF statement for obese type 2 diabetes. Diabet Med. 2011;28(6):628–42. 3. Longitudinal Assessment of Bariatric Surgery, C, et al. Perioperative safety in the longitudinal assessment of bariatric surgery. N Engl J Med. 2009;361(5):445–54. 4. DeMaria EJ, Portenier D, Wolfe L. Obesity surgery mortality risk score: proposal for a clinically useful score to predict mortality risk in patients undergoing gastric bypass. Surg Obes Relat Dis. 2007;3(2):134–40. 5. Finks JF, et al. Predicting risk for serious complications with bariatric surgery: results from the Michigan bariatric surgery collaborative. Ann Surg. 2011;254(4):633–40. 6. Higgins RM, et al. Preoperative immobility significantly impacts the risk of postoperative complications in bariatric surgery patients. Surg Obes Relat Dis. 2018;14(6):842–8. 7. Sharma G, et al. Comparative outcomes of bariatric surgery in patients with impaired mobility and ambulatory population. Obes Surg. 2018;28(7):2014–24. 8. Lavie CJ, Milani RV, Ventura HO. Obesity and cardiovascular disease: risk factor, paradox, and impact of weight loss. J Am Coll Cardiol. 2009;53(21):1925–32. 9. Samanta R, et al. Influence of BMI on inducible ventricular tachycardia and mortality in patients with myocardial infarction and left ventricular dysfunction: the obesity paradox. Int J Cardiol. 2018;265:148–54. 10. Litwin SE. Which measures of obesity best predict cardiovascular risk? J Am Coll Cardiol. 2008;52(8):616–9. 11. Poirier P, et al. Cardiovascular evaluation and management of severely obese patients undergoing surgery: a science advisory from the American Heart Association. Circulation. 2009;120(1):86–95. 12. Grandinetti A, et al. Association of increased QTc interval with the cardiometabolic syndrome. J Clin Hypertens (Greenwich). 2010;12(4):315–20. 13. Wijeysundera DN, et al. Risk of elective major noncardiac surgery after coronary stent insertion: a population-based study. Circulation. 2012;126(11):1355–62. 14. Oscarsson A, et al. To continue or discontinue aspirin in the perioperative period: a randomized, controlled clinical trial. Br J Anaesth. 2010;104(3):305–12. R. M. Tholey and D. S. Tichansky 15. Aminian A, et al. Who should get extended thromboprophylaxis after bariatric surgery?: a risk assessment tool to guide indications for post-discharge pharmacoprophylaxis. Ann Surg. 2017;265(1):143–50. 16. Finks JF, et al. Predicting risk for venous thromboembolism with bariatric surgery: results from the Michigan bariatric surgery collaborative. Ann Surg. 2012;255(6):1100–4. 17. Thereaux J, et al. To what extent does posthospital discharge chemoprophylaxis prevent venous thromboembolism after bariatric surgery?: results from a nationwide cohort of more than 110,000 patients. Ann Surg. 2018;267(4):727–33. 18. Karaman K, et al. Porto-mesenteric venous thrombosis after laparoscopic sleeve gastrectomy: a case report and systematic review of the 104 cases. Obes Res Clin Pract. 2018;12(3):317–25. 19. Birkmeyer NJ, et al. Preoperative placement of inferior vena cava filters and outcomes after gastric bypass surgery. Ann Surg. 2010;252(2):313–8. 20. Rowland SP, et al. Inferior vena cava filters for prevention of venous thromboembolism in obese patients undergoing bariatric surgery: a systematic review. Ann Surg. 2015;261(1):35–45. 21. Lopez PP, et al. Prevalence of sleep apnea in morbidly obese patients who presented for weight loss surgery evaluation: more evidence for routine screening for obstructive sleep apnea before weight loss surgery. Am Surg. 2008;74(9):834–8. 22. de Raaff CAL, et al. Perioperative management of obstructive sleep apnea in bariatric surgery: a consensus guideline. Surg Obes Relat Dis. 2017;13(7):1095–109. 23. Chung F, Abdullah HR, Liao P. STOP-bang questionnaire: a practical approach to screen for obstructive sleep apnea. Chest. 2016;149(3):631–8. 24. Wolter S, et al. Upper gastrointestinal endoscopy prior to bariatric surgery-mandatory or expendable? An analysis of 801 cases. Obes Surg. 2017;27(8):1938–43. 25. Abd Ellatif ME, et al. Place of upper endoscopy before and after bariatric surgery: a multicenter experience with 3219 patients. World J Gastrointest Endosc. 2016;8(10):409–17. 26. Salama A, et al. Is routine preoperative esophagogastroduodenoscopy screening necessary prior to laparoscopic sleeve gastrectomy? Review of 1555 cases and comparison with current literature. Obes Surg. 2018;28(1):52–60. 27. Goitein D, et al. Barium swallow for hiatal hernia detection is unnecessary prior to primary sleeve gastrectomy. Surg Obes Relat Dis. 2017;13(2):138–42. 28. Bennett S, et al. The role of routine preoperative upper endoscopy in bariatric surgery: a systematic review and meta-analysis. Surg Obes Relat Dis. 2016;12(5):1116–25. 29. Miller-Matero LR, et al. To eat or not to eat; is that really the question? An evaluation of problematic eating behaviors and mental health among bariatric surgery candidates. Eat Weight Disord. 2014;19(3):377–82. 30. Benzerouk F, et al. Food addiction, in obese patients seeking bariatric surgery, is associated with higher prevalence of current mood and anxiety disorders and past mood disorders. Psychiatry Res. 2018;267:473–9. 31. Steffen KJ, et al. Alcohol and other addictive disorders following bariatric surgery: prevalence, risk factors and possible etiologies. Eur Eat Disord Rev. 2015;23(6):442–50. 32. Madan AK, Tichansky DS, Taddeucci RJ. Postoperative laparoscopic bariatric surgery patients do not remember potential complications. Obes Surg. 2007;17(7):885–8. 33. Ronellenfitsch U, et al. The effect of clinical pathways for bariatric surgery on perioperative quality of care. Obes Surg. 2012;22(5):732–9. 8 Anesthetic Considerations Hendrikus J. M. Lemmens, John M. Morton, Cindy M. Ku, and Stephanie B. Jones Chapter Objectives 1. Provide the anesthesiologist with a practical approach to the problems that require special consideration in patients with severe obesity. 2. Identify comorbidities that require preoperative evaluation. 3. Evaluate anesthetic and analgesic options. 4. Discuss how obesity affects airway management and how bariatric surgery affects fluid management. 5. Review postoperative considerations. Introduction Anesthetic management of the patient with severe obesity (SO) presenting for bariatric surgery differs significantly from that of the normal-weight patient undergoing similar procedures. Well-planned and rational management of patients undergoing bariatric surgery requires detailed knowledge of how severe obesity affects anesthesia care. The mechanical effects of increased body mass index (BMI) as well as the physiological changes and associated comorbidities all impact safe management and decision-making by the anesthesiologist. The aim of this chapter is to provide a practical approach to the problems that require special consideration in patients with severe obesity. H. J. M. Lemmens Multispecialty Division, Department of Anesthesia, Stanford University School of Medicine, Stanford, CA, USA J. M. Morton Bariatric and Minimally Invasive Surgery Division, Yale School of Medicine, New Haven, CT, USA C. M. Ku · S. B. Jones (*) Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA e-mail: sbjones@bidmc.harvard.edu Preoperative Evaluation A full assessment for medical conditions that can affect perioperative complications must be performed. Preoperative “clearance” by a primary care physician or allied health professional and surgical consultation notes may not include information pertinent to anesthesia care, and while electronic health records can be helpful in information sharing, errors and gaps in information may be perpetuated. Organ dysfunction identified in the preoperative evaluation should be thoroughly evaluated and optimized before proceeding with this elective surgery. espiratory Issues Relevant to Anesthesia R Management The effect of obesity on the respiratory system decreases the margin of safety of anesthetic agents and increases the risk of respiratory failure in the perioperative period [1]. Respiratory failure is a life-threatening complication after bariatric surgery, with a reported incidence of 1.35–8% [2, 3]. Risk factors that increase the likelihood of respiratory failure are congestive heart failure, open surgery, chronic renal failure, peripheral vascular disease, male gender, age >50 years, alcohol abuse, chronic lung disease, diabetes, and smoking [2]. The presence of metabolic syndrome significantly increases the odds of postoperative pulmonary adverse events [4, 5]. At baseline, subjects with severe obesity may be mildly hypoxemic, with higher respiratory rates and lower tidal volumes. The compliance of the respiratory system is reduced and work of breathing increased. Functional residual capacity (FRC) and expiratory reserve volume (ERV) decrease exponentially with increasing BMI, with the greatest rate of change in the overweight (BMI 25.0–29.9 kg/m2) and mildly obese (BMI 30.0–34.9 kg/m2). In sitting subjects with a BMI of 30 kg/m2, FRC and ERV are 75 and 47%, respectively, of the values for a person with a BMI of 20 kg/m2 [6]. In anes- © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_8 89 90 will assist in that regard. Strategies such as reverse Trendelenburg positioning (when appropriate for the surgical procedure), periodic intraoperative lung recruitment, avoiding reabsorption atelectasis by keeping FiO2 <0.8, using less than 8 ml/kg of LBW for volume-controlled mechanical ventilation to avoid volutrauma, and utilizing incentive spirometry and noninvasive ventilation in the PACU may help prevent respiratory complications in the patient with obesity. ardiovascular Issues Relevant to Anesthesia C Management 80 70 60 50 Blood Volume (mL/kg) 90 100 The increased tissue mass of the patient with severe obesity needs to be perfused, leading to an increased total blood volume. While the total blood volume is increased, on a per kg total body weight basis, blood volume is decreased. Blood volume per kg total body weight (TBW) decreases exponentially from 70 ml/kg TBW at a BMI of 22 kg/m2 to 40 ml/kg TBW at a BMI of 65 kg/m2 (Fig. 8.1) [16]. If 70 ml/kg TBW is used for blood volume calculation, blood volume is overestimated, and under-transfusion of blood products may occur when significant blood loss during surgery is encountered. The increased total blood volume of the patient with SO results in an increased cardiac output (CO). CO increases from 4 L/min at a BMI of 20 kg/m2 to more than 6 L/min at BMIs greater than 40 kg/m2. CO affects early pharmacokinetics, the front-end kinetics of drug distribution, and dilution in the first minutes after administration. An increased CO decreases the fraction of drug distributed to the brain and increases the rate of redistribution, which will result in lower 40 thetized supine patients, the effect of BMI on FRC is more pronounced, and tidal volume is more likely to fall within closing capacity, promoting shunting [7]. In addition to the changes in respiratory mechanics and lung volumes, the prevalence of obstructive sleep apnea (OSA) in bariatric patients can be as high as 75% [8]. Of those with OSA and severe obesity, up to 20% may have obesity hypoventilation syndrome (OHS), which is characterized by daytime awake hypercapnia, hypoxemia, and elevated HCO3− [9]. It is important for anesthesiologists to recognize patients with OHS since it is associated with severe upper airway obstruction, restrictive lung disease, blunted central respiratory drive, pulmonary hypertension, and increased mortality. Before bariatric surgery, these patients should be referred to sleep medicine for polysomnography and positive airway pressure (PAP) titration. An echocardiogram should be performed to assess right ventricular function and pulmonary hypertension. Poor right ventricular function and high mean pulmonary artery pressures (>35 mmHg) are associated with an unacceptable anesthesia-related perioperative mortality risk. Perioperative precautions for OHS include prudent airway management, rapid emergence, monitoring for ventilatory impairment, and early resumption of PAP therapy. The compromised respiratory status of the patient with obesity requires special precautions to prevent oxygen desaturation at induction of anesthesia, during surgery, and in the postoperative phase. After preoxygenation with 100% oxygen and complete denitrogenation, an anesthetized supine paralyzed SO patient’s oxygen saturation decreases from 100% to 90% in about 2.5 min [10]. Premedication with anxiolytics or opioids depresses spontaneous ventilation and should be minimized. Preoxygenation techniques delaying the onset of hypoxia should be used: the 25° head-up position may add a minute to the safe apnea time [11], the use of supplemental oxygen or high flow oxygen decreases the rate of desaturation [12], and application of noninvasive positive airway pressure during induction of anesthesia can increase safe apnea time by 50% [13] while improving oxygenation during maintenance of anesthesia [14]. Immediately after induction of anesthesia, atelectasis develops mainly in the dependent areas of the lung. Atelectasis results in pulmonary shunting and hypoxemia. Release of inflammatory cytokines associated with atelectasis may contribute to postoperative ventilator-associated lung injury such as pneumonia and respiratory failure. In the patient with severe obesity, ventilatory strategies to prevent atelectasis may require recruitment maneuvers and positive end-expiratory pressure (PEEP) values higher than 10 cm H2O. Although high PEEP levels combined with pneumoperitoneum will decrease venous return and can induce hypotension, PEEP levels up to 20 cm H2O are generally tolerated in wellhydrated patients [15]. The trend toward maintaining preoperative normovolemia as part of enhanced recovery protocols H. J. M. Lemmens et al. 20 30 40 50 Body Mass Index (kg/m2) 60 70 Fig. 8.1 Effect of BMI on blood volume in mL per kg TBW. Blood volume (mL per kg TBW) for a patient with a given BMI (BMIp) can be calculated using the equation: 70 BMIp 22 Pharmacological Considerations Until recently, obese subjects have been routinely excluded from clinical trials to obtain regulatory approval for investigational drugs. This has resulted in package insert dosage recommendations based on total body weight, valid for normal-­weight patients but not for the obese. Severe obesity alters the pharmacokinetics and drug response of anesthetic agents. In addition, the decreased pulmonary and cardiac reserve of the patient with SO significantly decreases the margin of safety of anesthetic agents. Incorrect dosing can increase the rate of perioperative complications. Obesity is not only associated with an increase in tissue mass but also changes in body composition and tissue perfusion. Fat mass and lean body mass both increase, but the increase is not proportional. The percentage of lean body mass as a percentage of total body weight decreases as BMI increases (Fig. 8.2). The different ratio of lean body weight (LBW) to fat weight at different BMIs has a significant impact on drug distribution. Fat perfusion is also altered at different BMIs. At low BMIs, fat is relatively well perfused; at high BMIs fat is poorly perfused. Because of the different ratio of fat to lean body weight at different BMIs and changes in fat perfusion, the effect of obesity on drug distribution into the different tissues is poorly understood. The increased CO of the patient with SO increases the dose requirements of induction agents, but not to the level of total body weight. In patients with normal cardiac function, CO is Kg 100 150 Total Body Weight Lean Body Weight 50 concentrations, faster awakening, and increased dose requirement. This phenomenon has important implications for the induction dose of intravenous anesthetic agents. The most prevalent comorbidity in bariatric patients is hypertension. Obesity may lead to abnormal cardiac function through pathways that are associated with or independent of hypertension. The mechanisms of decreased cardiac contractility associated with obesity independent of hypertension are related to metabolic dysregulation, but not completely understood. The increased body mass, metabolic syndrome, insulin resistance, type 2 diabetes, and physical inactivity all contribute to systolic and diastolic dysfunction even in otherwise healthy young obese subjects. This may eventually progress to left and/or right heart failure. The combination of super obesity (BMI > 50 kg/m2) with hypertension and diabetes is associated with a twofold increased risk of death and adverse cardiac events in the perioperative phase [5]. Congestive heart failure, peripheral vascular disease, and chronic renal failure are predictive factors of increased in-­ hospital mortality after surgery. Obesity is also associated with an increased risk of atrial fibrillation and ventricular ectopy. Cardiac events can be a significant cause of 30-day mortality after bariatric surgery. 91 200 Anesthetic Considerations 0 8 Fat Weight 20 30 40 50 60 70 BMI (kg/m2) Fig. 8.2 Changes in body composition for a typical frame 160-cm-tall female who increases her BMI. Lean body weight was calculated using the equations published by Janmahasatian et al. [17]. Fat weight was calculated by subtracting lean body weight from total body weight highly correlated to lean body weight, more so than total body weight or other variables. Therefore, lean body weight and CO are more appropriate dosing scalars than total body weight. Total body weight dosing of induction agents will result in overdosing and side effects such as hypotension. Numerous pharmacokinetic studies have shown that clearance, the most relevant pharmacokinetic parameter for maintenance dosing, is linearly related to lean body weight but not total body weight. This implies that lean body weight is the appropriate dosing scalar, not only for determining induction and loading doses but also for maintenance doses. Recommended dosing scalars for several anesthetic agents are summarized in Table 8.1. Induction Agents Thiopental Although thiopental is part of the World Health Organi­ zation’s “Essential Drugs List,” it is currently unavailable in the United States. Nevertheless, thiopental is an ideal anesthetic induction agent with arguably less side effects than propofol. Immediately after intravenous (IV) administration, thiopental distributes to highly perfused tissues such as the brain, lung, liver, heart, kidney, gut, and pancreas. After an anesthetic induction dose, redistribution into the muscle depletes thiopental from the brain and terminates the anesthetic effect within 5–10 min. The increased CO associated with severe obesity has a significant effect on the thiopental dose requirement. After a thiopental induction dose of 250 mg, the higher CO of a patient with severe obesity results in peak arterial concentrations up to 50% lower than those of 92 H. J. M. Lemmens et al. Table 8.1 Recommended dosing scalars for morbidly obese patients Dosing scalar Induction agents Thiopental LBW Propofol LBW Etomidate LBW Opioids Fentanyl Alfentanil Sufentanil Remifentanil LBW LBW LBW LBW Muscle relaxants Succinylcholine TBW Rocuronium Vecuronium Cisatracurium Atracurium LBW/ IBW LBW/ IBW LBW/ IBW LBW/ IBW Comments For continuous infusion or maintenance dosing TBW Use in septic patients is controversial; may cause adrenal suppression Titrate to effect TBW dosing may result in significant hypotension and/or bradycardia The incidence of myalgia is low in morbidly obese patients IBW dosing results in shorter duration of action Fast administration may result in histamine release LBW lean body weight, TBW total body weight, IBW ideal body weight a lean subject. Thiopental dose adjusted according to lean body mass or increased CO results in the same peak plasma concentration as for a normal size person. Propofol In current practice, propofol is the induction agent of choice for obese patients. CO has a significant effect on peak plasma concentration and duration of effect. After a bolus dose for induction of anesthesia, propofol’s peak plasma concentration is inversely related to CO. In addition, a higher CO is associated with a faster wake-up time. For COs of 8.5, 5.5, and 2.5 L/min, recovery of consciousness is predicted to occur at 2.9, 8.6, and 18.7 min, respectively. CO does not affect onset time. LBW is a more appropriate weight-based scalar than TBW for propofol induction of general anesthesia in patients with SO [18]. Patients in whom anesthesia was induced with propofol dose based on LBW required similar doses of propofol and had similar times to loss of consciousness compared to nonobese control patients given propofol based on TBW. Etomidate Etomidate is less likely to cause a significant decrease in blood pressure than thiopental or propofol. Thus, in patients with significant heart disease or hemodynamically unstable patients, anesthetic induction with etomidate may be a better choice. The pharmacology of etomidate in obese patients has not been studied, but an induction dose based on lean body mass and CO can be justified given the pharmacokinetic and pharmacodynamic similarities of etomidate, thiopental, and propofol. Etomidate transiently suppresses corticosteroid synthesis in the adrenal cortex by reversibly inhibiting 11-beta-­ hydroxylase. This suppressant effect on steroid synthesis is probably clinically insignificant after a single dose used for induction of anesthesia. However, in patients with sepsis, the use of etomidate for induction of anesthesia is controversial. Other side effects are pain at injection, myoclonus, and a high incidence of postoperative nausea and vomiting. Dexmedetomidine Dexmedetomidine is used as a sedative agent with both anxiolytic and analgesic effects. Dexmedetomidine is a selective alpha-2 adrenergic receptor agonist. Respiratory depression is minimal, but dexmedetomidine potentiates the respiratory depressant effect of opioids and benzodiazepines. The short distribution half-life (8 min) and relatively short elimination half-life (2 h) make it suitable for titration by continuous infusion. The sympatholytic effect of dexmedetomidine decreases norepinephrine release and will decrease arterial blood pressure and heart rate. This may result in severe hypotension in hypovolemic patients and severe bradycardia in patients with heart block. Another side effect is dry mouth, which when used during fiber-optic intubation is an advantage. Postoperatively, dexmedetomidine reduces shivering. During open gastric bypass surgery when used instead of fentanyl to supplement desflurane, a loading dose of dexmedetomidine, 0.5 mcg/kg, given over 10 min followed by an infusion of 0.4 mcg/kg/h, resulted in significantly lower arterial blood pressure and heart rate, shorter time to tracheal extubation, lower pain scores, and less use of morphine and antiemetics in the postanesthesia care unit (PACU). A ­systematic review of 30 studies with intraoperative use of dexmedetomidine showed a decrease in morphine consumption, lower pain scores, and lower rates of PONV [19]. For adjunctive use during laparoscopic bariatric surgery, a lower infusion rate (0.2 mcg/kg/min) is recommended to reduce the risk of cardiovascular side effects. Opioids Compared to fentanyl and its analogues alfentanil, sufentanil, and remifentanil, the longer-acting opioids morphine and hydromorphone are not potent enough to effectively 8 Anesthetic Considerations block somatic and autonomic responses during surgery. In addition, the drowsiness and sleepiness at emergence associated with morphine and hydromorphone administration are unwanted in the morbidly obese. Therefore, their use during surgery is best avoided. If necessary, morphine should be dosed with IBW [20] and titrated to effect with close respiratory monitoring, as opioid-related adverse events remain a significant complication in SO patients [21]. Fentanyl Fentanyl has a fast onset of effect (5 min) and effectively blocks somatic and autonomic responses during surgery. Fentanyl is probably the most commonly used opioid during bariatric surgery. The higher CO in patients with obesity will result in significantly lower fentanyl concentrations in the early phase of distribution. Pharmacokinetic parameters of normal size persons will overpredict measured fentanyl concentrations in patients with obesity. The clearance of fentanyl is higher in patients with obesity and increases nonlinearly with increasing TBW, but linearly with lean body weight (LBW). These data suggest loading and maintenance doses of fentanyl should be based on LBW. However, obesity increases the probability of respiratory depression in the perioperative period, and fentanyl and other opioid administration should be carefully titrated according to individual patient’s need. Sufentanil Sufentanil is highly lipophilic and has an onset time of approximately 5 min. Like fentanyl, pharmacokinetic parameters of normal size persons will overpredict measured sufentanil concentrations in patients with severe obesity. Alfentanil Alfentanil has a fast-onset time of approximately 1 min. The higher CO in patients with obesity will result in significantly lower alfentanil concentrations in the early phase of distribution. Alfentanil is less lipid soluble than fentanyl or sufentanil and has a smaller volume of distribution. No data on the effects of obesity on the pharmacokinetics of alfentanil have been published. Remifentanil Remifentanil’s physicochemical properties result in a rapid-­onset time of approximately 1 min. Bolus admin- 93 istration in awake patients may result in severe bradycardia, hypotension, and muscle rigidity. Plasma and tissue esterases rapidly hydrolyze remifentanil, resulting in an extraordinary high clearance (3 L/min) unaffected by hepatic or renal insufficiency. The fast-onset time and high clearance make remifentanil especially suitable for administration by continuous infusion. Volumes and clearances not normalized for weight are similar in obese and nonobese patients and do not correlate with TBW, but correlate significantly with LBW. Therefore, in the patient with obesity, dosing of remifentanil based on TBW will result in concentrations higher than those needed for clinical purposes and an increased incidence of side effects such as hypotension and bradycardia. Remifentanil dosing based on LBW will result in plasma concentrations similar to those in normal-weight subjects when dosed according to TBW. After discontinuation of administration, drug effects terminate rapidly, within 5–10 min. Therefore, when postoperative pain is anticipated, alternative analgesics should be administered prior to remifentanil’s discontinuation. Remifentanil may also induce postoperative hyperalgesia, requiring higher than normal dosages of longer-acting opioids to effectively treat pain, putting the patient with severe obesity at higher risk of opioid complications such as respiratory depression. Inhaled Anesthetics Isoflurane The solubility of inhaled anesthetic agents in fat and the increased fat mass of the patient with obesity would theoretically result in an increased anesthetic uptake, especially with more fat-soluble anesthetics such as isoflurane. However, blood flow per kg of fat tissue decreases significantly with increasing BMI, therefore limiting uptake. In addition, the time constants (the time to reach 63% of equilibrium) for equilibrium with fat are long (2110 and 1350 min for ­isoflurane and desflurane, respectively). The decreased fat perfusion and relatively long time constants will diminish the effect of the increased fat mass on the uptake of inhalational agents. During routine clinical practice, the effect of BMI on the uptake of desflurane and the more lipid-soluble isoflurane is clinically insignificant. The concern that isoflurane prolongs emergence from anesthesia in patients with obesity due to its lipid solubility could also not be substantiated. Obese and nonobese patients emerged from anesthesia at similar times (7 min) after 0.6 MAC isoflurane administration for procedures lasting 2–4 h. After termination of isoflurane administration, the time to extubation can be decreased significantly by increasing alveolar ventilation using an isocapnic hyperpnea method [22]. 94 Desflurane The effect of BMI on desflurane uptake is insignificant, and obese and nonobese patients emerge from anesthesia equally rapidly (4 min) after 0.6 MAC desflurane administration for procedures lasting 2–4 h. Several studies in patients with obesity have compared desflurane and sevoflurane with variable results, finding either a faster awakening with desflurane or no difference [23, 24]. Sevoflurane Sevoflurane appears to provide a slightly more rapid uptake and elimination of anesthetic in patients with severe obesity than does isoflurane. Fluoride, a metabolite of sevoflurane, in concentrations greater than 50 mmol/L, can be nephrotoxic. In addition, sevoflurane is degraded to compound A by carbon dioxide absorbers containing a strong base such as barium hydroxide lime or to a lesser extent by soda lime. Reductions in fresh gas flow as well as an increase in temperature in the gas mixture will increase compound A concentrations. Albuminuria, glycosuria, and enzymuria are associated with inhaled doses of compound A greater than 160 ppm/h. In the few studies in patients with renal impairment, no evidence of further worsening of renal function could be demonstrated after sevoflurane administration. However, the safety of sevoflurane in patients with impaired renal function is unclear. Neuromuscular Blocking Agents Succinylcholine Succinylcholine is a depolarizing muscle relaxant. It is a nicotinic acetylcholine receptor agonist causing fasciculations followed by flaccid paralysis via depolarization of the motor end plate. Succinylcholine has the fastest onset and shortest duration of action of all muscle relaxants— excellent properties to achieve intubation of the trachea rapidly. If difficulty is encountered managing the airway of the patient, return of neuromuscular function and spontaneous ventilation will occur within 5–7 min. Maximum effect and duration of action are determined by the extracellular fluid volume and elimination by the plasma enzyme butyrylcholinesterase (also known as pseudocholinesterase). Extracellular fluid volume and activity of butyrylcholinesterase both increase with increasing BMI. Therefore, patients with severe obesity have larger succinylcholine requirements than normal size patients. Succinylcholine, 1 mg/kg total body weight, will result in complete neuromuscular blockade and excellent intuba- H. J. M. Lemmens et al. tion conditions in the obese. Lower doses are associated with poor intubating conditions due to incomplete neuromuscular block. Succinylcholine use is associated with increases in potassium and myalgia. The incidence of succinylcholine-induced myalgia is low in morbidly obese patients. Rocuronium Rocuronium is a nondepolarizing muscle relaxant that can be used as an alternative to succinylcholine for rapid sequence intubation. A dose of 1.2 mg/kg ideal body weight (IBW) provides excellent or good intubating conditions 60 s after administration. However, the time to reappearance of T1 is 52 min. Rocuronium maintenance dosing based on lean body weight has not been studied, but dosing on the basis of ideal body weight is appropriate. Maximum effect and recovery times of rocuronium and all other muscle relaxants are highly variable; therefore, continuous monitoring of the degree of neuromuscular blockade is recommended. The use of rocuronium for rapid sequence intubation has become more common with the wider availability of sugammadex (below) for urgent reversal, if needed. Vecuronium The pharmacokinetic parameters uncorrected for weight are similar between obese and nonobese subjects. In obese subjects receiving 0.1 mg/kg vecuronium based on TBW, recovery times from neuromuscular blockade were approximately 60% longer than in normal-weight control subjects. The prolonged recovery from neuromuscular blockade in the obese patients can be explained by the larger dose of vecuronium. The similar pharmacokinetics will result in higher vecuronium plasma concentration in the patient with obesity. With higher doses and higher plasma concentrations, ­recovery from neuromuscular blockade will occur at a time when plasma concentration decreases more slowly, instead of the more rapid decline after a smaller dose when recovery occurs earlier during the distribution phase. To avoid overdose in the patient with obesity, administering vecuronium on the basis of IBW is recommended. Cisatracurium Cisatracurium is eliminated via Hoffman degradation, a pathway independent from kidney or liver function. As expected, TBW dosing results in a prolonged duration of action when compared with a control group of normal body weight patients. When administered to patients with severe 8 Anesthetic Considerations obesity based on IBW, the duration of action of cisatracurium was decreased when compared to normal size patients. 95 Monitoring In the relatively healthy patient undergoing bariatric surgery, noninvasive monitoring during anesthesia will suffice. There Reversal Agents of Neuromuscular Blockade are no data showing that invasive monitoring improves outcome in patients with severe obesity without advanced carRapid and complete recovery from neuromuscular blockade diac or pulmonary disease [28]. is particularly important in the morbidly obese patient. During surgery, a combination of lead II and lead V5 elecResidual neuromuscular blockade will further compromise trocardiographic monitoring has a sensitivity of 80% to respiratory function in the immediate postoperative phase. detect myocardial ischemia. Lead V4 and V5 monitoring has Obesity is a risk factor for residual blockade [25]. a sensitivity of 90%. The best combination is lead V4, V5, Acceleromyography should be used preferentially over tra- and II monitoring, resulting in a sensitivity of 98%. Cardiac ditional twitch monitoring when monitoring and reversing abnormalities such as rhythm and conduction problems neuromuscular blockade [26] and has been shown to signifi- occur frequently in the patient with severe obesity. Atrial cantly reduce the incidence of residual block. fibrillation is the most commonly occurring abnormal rhythm, especially in patients with OSA. If during surgery or in the postoperative phase atrial fibrillation develops, atrial Neostigmine distension due to fluid overload can be the causative factor. Prolonged QT interval syndrome is a precursor of torsades The dose-response relationship of neostigmine for neuro- de pointes, which can result in sudden cardiac death. Many muscular blockade reversal in morbidly obese patients has drugs used in the perioperative phase such as ondansetron, been poorly studied. When vecuronium is reversed with neo- sevoflurane, and methadone prolong the QT interval and stigmine at 25% recovery of twitch height, there is no differ- should not be used in patients with prolonged QT interval ence between normal size and morbidly obese patients in syndrome. A well-fitting blood pressure cuff encircling at least 75% time to a recovery of train-of-four (TOF) ratio to 0.7 (3.8– 4.8 min). However, recovery time to adequate reversal (a of the arm should be used to obtain reliable blood pressure train-of-four ratio of 0.9) is four times slower in morbidly measurements. A blood pressure cuff that is too large will obese patients (25.9 versus 6.9 min). The recommended dose underestimate blood pressure. A cuff that is too small will of neostigmine is 0.04–0.08 mg/kg, not to exceed a total dose overestimate blood pressure. Special conical-shaped cuffs of 5 mg. A deep neuromuscular block (TOF ratio of 0) can- for morbidly obese patients are available. If a blood pressure cuff cannot be fitted on the upper arm, a standard blood presnot be reversed with neostigmine. sure cuff placed on the forearm is a useful alternative. Lower arm measurements overestimate blood pressure. The threshold to place an intra-arterial catheter should be low since Sugammadex invasive blood pressure measurement is accurate and comSugammadex is the first selective relaxant-binding agent plications of radial arterial line placement are rare. The recommendation that a central venous line be inserted specifically designed to bind and encapsulate rocuronium and vecuronium. It can provide immediate reversal of an routinely in obese patients is not valid. Peripheral venous intubating dose of rocuronium at 16 mg/kg. The muscle access can be more difficult, but this is not a definitive indirelaxant is bound with high affinity within sugammadex’s cation for a central line. If placement of a peripheral venous core and cannot bind to the acetylcholine receptor at the neu- catheter is problematic, ultrasound can be used to locate a romuscular junction. The bound complex is excreted by the peripheral vein. Central venous access via the internal jugukidneys at a rate equal to the glomerular filtration rate. Unlike lar vein is associated with a lower complication rate than the acetylcholinesterase inhibitor neostigmine, sugammadex subclavian vein puncture. Positioning the patient on a ramp has no effect at the receptor level, and there is no need to similar to the positioning used for tracheal intubation with a coadminister antimuscarinic agents such as atropine or gly- roll under the shoulders will maximize neck exposure and copyrrolate. Deep neuromuscular blockade (TOF =0, post-­ facilitate placement. Thereafter, the patient can be placed in tetanic count of 1–2) can be reversed with 4 mg/kg Trendelenburg position as tolerated. Insertion of a central sugammadex at IBW [27], and at a moderate level of block- venous catheter under ultrasound guidance facilitates correct placement and is currently the recommended approach [29]. ade (TOF = 2), 2 mg/kg is recommended. The value of the central venous pressure (CVP) measureSugammadex was approved for use in the European Union in 2008 and subsequently approved by the US Food ment does not necessarily reflect adequacy of circulating blood volume or response to fluid loading. A decreased CVP and Drug Administration (FDA) in 2015. 96 can reflect venodilation or hypovolemia. An increased CVP can reflect decreased cardiac pump function, increase in thoracic pressure and/or pericardial pressure, or increased pulmonary artery resistance. In contrast to the CVP pressure readings, the shape of the CVP waveform can be highly diagnostic. For example, the presence of large v waves, which are diagnostic for tricuspid regurgitation, may indicate the presence of pulmonary hypertension and right heart failure. Intravascular volume status is difficult to assess in obese patients. Pulse pressure variation, the decrease in arterial pulse pressure with positive pressure ventilation, is a more reliable indicator of hypovolemia than CVP. During inspiration of controlled ventilation, the great veins entering the heart are compressed, resulting in a reduction of right ventricle preload and an increase in afterload. The decreased preload and increased afterload decrease the stroke volume of the left ventricle at the end of the expiration cycle. When this is exaggerated, the blood volume of the patient is contracted, and a fluid bolus will improve CO. Commercial devices are being marketed that provide real-time changes in pulse pressure variation. However, these devices typically require positive pressure ventilation and higher tidal volumes, which may be contrary to the goals of lung protective ventilation strategies. Pulmonary artery pressure monitoring has fallen into disfavor because it is a poor indicator of left ventricle preload or circulating blood volume, but it is still being used to assess pulmonary hypertension. Transesophageal echocardiography (TEE) is an invaluable tool to assess the possible cause of sudden intraoperative cardiac or hemodynamic instability such as myocardial ischemia associated with wall motion abnormalities, emboli, and hypovolemia. TEE cannot be used routinely during bariatric surgery because the ultrasound probe is in the surgical field. The degree of neuromuscular blockade should be monitored during surgery. During surgery, train-of-four monitoring (TOF) monitoring is used to guide the administration of neuromuscular blocking agents. Acceleromyography should be utilized over traditional qualitative twitch monitors, if available. At the end of surgery, the TOF ratio is used to guide administration of reversal agents. A deep neuromuscular block (TOF ratio of 0) cannot be reversed with neostigmine, and reversal should be delayed until a TOF count of 2 is observed. The administered dose of neostigmine should not exceed 5 mg, and the trachea should not be extubated with a TOF ratio of <0.9. As noted above, deep neuromuscular blockade can be reversed with sugammadex at 4 mg/kg IBW. Electroencephalographic-based brain function monitors are being used to guide administration of anesthetic agents and to prevent awareness. The efficacy of these monitors is H. J. M. Lemmens et al. controversial, and routine use of brain function monitoring during surgery is not supported by data. However, its use is recommended in cases where a high risk for awareness may be suspected. Administration of nitrous oxide, ketamine, isoflurane, or halothane can be associated with a paradoxical increase in BIS. A Foley urinary catheter may be placed before any major surgery, but not required if the surgery is anticipated to be of short duration and routine. A low urine output may be encountered during laparoscopic bariatric surgery. Pneumoperitoneum decreases urine output by increasing ADH, aldosterone, and plasma renin activity. Preoperative Sedation For most patients, premedication with sedatives should be used with caution. Respiratory depression caused by benzodiazepines and opioids is pronounced in patients with severe obesity, especially in those with OSA. The upper airway collapsibility of the patient with severe obesity combined with the decreased arousal response to airway occlusion makes these patients particularly sensitive to drug-induced respiratory depression. Benzodiazepines decrease upper airway muscle activity with consequent obstruction and cause central apnea during the initial post-administration minutes. If very anxious patients need premedication, small doses of midazolam can be administered upon transport to the operating room under continued visual monitoring and verbal communication during transport. Administration of oxygen during transport is recommended. In the operating room, it is practical to have unpremedicated patients climb off the gurney and then position themselves on the operating room table. Airway Management According to recent data from the American Society of Anesthesiologists Closed Claim Database, almost 40% of adverse airway events during induction of anesthesia occurred in obese patients, with a significant proportion resulting in permanent brain damage or death [30]. This is not surprising since inability to mask ventilate and intubate the morbidly obese patient will result in rapid development of hypoxemia. An understanding of the different requirements for airway management and adequate preparation is vital to avoid untoward events. The supine position, routinely used for induction of anesthesia in nonobese patients, is not appropriate for the patient with severe obesity. The supine position will decrease the FRC further and will increase the intra-abdominal pressure, 8 Anesthetic Considerations resulting in restriction of diaphragmatic movement with respiration. The torso of the morbidly obese should be elevated 25–30° prior to induction of anesthesia in reverse Trendelenburg position. Offsetting the mass loading of the abdomen and chest will increase the FRC, decrease the restrictive component of the lung function, and increase compliance. Therefore, reverse Trendelenburg positioning will not only improve the ability to ventilate the lungs by bag and mask but also will increase the time before desaturation starts to occur when there is an inability to manage the airway. The standard “sniffing position” for tracheal intubation, which results in optimal alignment of axes of the head and neck in the nonobese, is inadequate for the patient with severe obesity. A “ramped” position, with the patient’s upper body, head, and neck elevated until the external acoustic meatus and the sternal notch are in horizontal alignment, results in optimal position for tracheal intubation (Fig. 8.3). It also provides increased submandibular space facilitating manipulating the laryngoscope handle and laryngoscopy. Obesity itself is not an independent risk factor for difficult tracheal intubation, but patients with obesity plus a large neck circumference, excessive pretracheal adipose tissue, and high Mallampati classification do have a higher probability of difficult intubation. Obesity and OSA are independent risk factors for difficult mask ventilation. Since facial hair can make ventilation of the lungs by bag and mask ineffective, it is appropriate to ask patients to shave beards preoperatively. When very difficult mask ventilation and/or tracheal intubation is suspected, awake fiber-optic intubation (FOI) is the technique of choice. Spontaneous ventilation and airway patency will be maintained in the awake patient, assuming sedatives are carefully titrated—keeping in mind that even small amounts may result in airway obstruction. Adequate topical anesthesia of the larynx and pharynx and gentle technique will result in intubation with minimal discomfort for the patient. Visualization of the larynx may be difficult due to 97 airway narrowing by redundant folds of fat tissue. In a sitting patient, the pharyngeal space will be increased, and instructing the patient to protrude the tongue will further increase the diameter of the airway. Several reports have described the use of a laryngeal mask airway in an awake patient to open the surrounding tissues permitting easier visualization of the vocal cords and passage of the fiberscope. Video laryngoscopy, with multiple models available on the market, has become a ubiquitous and invaluable alternative to FOI and DL in the patient with severe obesity. The video laryngoscope allows for indirect visualization of the vocal cords and rapid intubation in the anesthetized patient. However, due to the indirect visualization, the tonsillar pillars and soft tissue in the pharynx and hypopharynx are not visualized well and are at risk for traumatic injury [31] during intubation; substantial training is required [32]. Aspiration Risk The traditional belief that patients with obesity are at higher risk for aspiration at induction of anesthesia is unfounded. The volume of gastric contents is not greater in fasted patient with obesity when compared to normal-weight subjects, and gastric emptying is not delayed, except in the setting of obesity-­related disease as noted below. In fact, the proportion of high-volume low pH gastric content in obese unpremedicated subjects is lower (26.6%) when compared to normal-weight patients (42%). Patients with comorbid conditions such as symptomatic gastroesophageal reflux disease, diabetes mellitus, and gastroparesis are at increased risk for gastric acid aspiration. A rapid sequence induction with cricoid pressure should be performed in these patients as well as in all patients with a prior gastric banding procedure. There are, however, emerging studies using biomarkers suggesting that cricoid pressure does not necessarily lead to decreased aspiration in these patients. Pneumoperitoneum: Implications for Ventilation, Hemodynamics, and Urine Output Fig. 8.3 Ramped position for tracheal intubation After insufflation of the abdomen, the increased intra-­ abdominal pressure and absorption of CO2 account for the majority of physiologic alterations. The diaphragm moves in the cephalad direction, which may result in migration of the endotracheal tube into the right main stem bronchus. High peak inspiratory pressures followed by hypoxemia and hypotension will result if this phenomenon is not recognized and corrected in a timely fashion. Appropriate ventilatory adjust- 98 H. J. M. Lemmens et al. ments can eliminate most of the absorbed CO2 and prevent Maintenance with balanced salt solutions such as lacacid-base disturbances. Reverse Trendelenburg position, tated Ringers is preferred, with boluses as needed when >10 cm PEEP, and recruitment maneuvers can dramatically signs and symptoms of hypovolemia present. Under anesimprove oxygenation in the morbidly obese in both open and thesia, use of a vasopressor such as phenylephrine may be a laparoscopic cases [33]. better choice than repeated fluid boluses, as decreased blood Upon insufflation, heart rate and blood pressure typically pressure is more likely due to decreased vascular tone rather increase; however, some patients may experience vagal-­ than hypovolemia. It is important to realize that heart rate, mediated bradycardia due to peritoneal stretching—a phe- blood pressure, urine output, and central venous pressure nomenon typically short-lived and relieved with peritoneal are not particularly accurate measures of volume status, desufflation and administration of IV atropine or glycopyrro- especially during laparoscopic procedures. Variations in late. In patients with severe obesity, CO decreases with insuf- stroke volume or pulse pressure can be utilized for goalflation, while systemic vascular resistance, pulmonary vascular directed fluid therapy, but these measurements are less accuresistance, mean arterial pressure, right atrial pressure, and rate in the presence of pneumoperitoneum and lower tidal pulmonary capillary wedge pressure increase. In general, the volume ventilation. In addition, benefits of goal-directed impact of pneumoperitoneum on the cardiovascular system of fluid therapy have not been as evident when the patient is the patient with severe obesity is well tolerated. euvolemic, as is more commonly the case for enhanced A decrease in renal perfusion occurs with pneumoperito- recovery pathways [35]. neum as well as increased release of antidiuretic hormone, plasma renin activity, and serum aldosterone, resulting in water retention and a rapidly decreasing urine output. The Postoperative Considerations extent of intraoperative oliguria is directly related to the extent of increased intra-abdominal pressure. Despite the In laparoscopic bariatric surgery, a deep neuromuscular hormonal changes and transient oliguria, clinically signifi- block is needed to ensure an adequate surgical field. After cant renal impairment as measured by serum creatinine does completion of surgery, before tracheal extubation is not occur after laparoscopic gastric bypass with insufflation attempted, it is imperative to completely reverse the neuropressures not exceeding 15 mmHg. However, in patients muscular block. Even minimal residual paralysis causes retwith preexisting renal impairment, minimal insufflation roglossal and retropalatal narrowing during inspiration, pressure and adequate IV hydration should be instituted to which may result in upper airway collapse. avoid further exacerbation of renal insufficiency. Use of continuous positive airway pressure (CPAP) Effects of pneumoperitoneum on hepatic blood flow and reduces the risk for atelectasis, and noninvasive ventilafunction are similar to those of the kidneys, and transaminase tion can be used as a prophylactic and/or therapeutic tool levels may rise as much as sixfold 24 h after surgery. These to improve gas exchange postoperatively. There is coneffects may be enhanced in morbidly obese patients because cern CPAP may increase the likelihood of an anastomotic many have underlying hepatic disease due to fatty leak by air forced into the gastric pouch. However, it has infiltration. been demonstrated that changes in transmural gastric Use of sequential compression devices (SCDs) in combi- pouch pressure with the application of CPAP do not occur nation with pneumoperitoneum is associated with a signifi- [36]. cant improvement in CO, stroke volume, portal venous and The American Society of Anesthesiologists (ASA) prachepatic arterial blood flow, and marked improvement in renal tice guidelines for the perioperative management of patients perfusion, urine output, and systemic vascular resistance. with OSA recommended that patients with OSA treated with CPAP should continue CPAP as soon as feasible after surgery [37]. The same guideline recommends that patients Fluid Management with OSA be monitored for 3 h longer than their non-OSA counterparts before discharge from the PACU. The recomGoals for fluid management should center around mainte- mendations for increased monitoring are based on expert nance of euvolemia, when possible. Extended fasting is opinion and not scientific evidence. The same guidelines avoided, and oral intake is resumed as soon as possible post- caution performing upper abdominal laparoscopic surgery in operatively [34]. The concept of the “third space” has been an outpatient setting for patients with known or suspected shown to be false; administered fluid either remains intravas- OSA. In a series of 746 patients with obstructive sleep apnea cular or distributes to the interstitial space. The increase in after ambulatory laparoscopic gastric banding, 40% did have hydrostatic pressure damages the endothelial glycocalyx and an incidence of hypoxia in either the OR or PACU [38]. alters vascular permeability, leading to gut wall edema and However, there were no episodes of respiratory failure or traileus. cheal reintubation. 8 Anesthetic Considerations Postoperative Nausea and Vomiting 99 sion. The laparoscopic approach provides substantial benefit with less postoperative pain and medication use. Bariatric surgery is associated with a high incidence of postPostoperative oral administration of analgesics can be utioperative nausea and vomiting (PONV). Besides the surgery lized in the ambulatory surgery setting, and oral absorption itself, female sex, a history of PONV, and motion sickness of drugs is essentially unchanged in obese patients, with liqare known risk factors. Opioids, volatile anesthetics, and uid preparation being best tolerated. The most common analnitrous oxide all have dose-related emetogenic effects. The gesic drugs given orally are nonsteroidal anti-inflammatory most commonly used antiemetic agent for the prevention of drugs (NSAIDs), acetaminophen, and opioids. NSAIDs are PONV is the serotonin antagonist ondansetron, with a half-­ not generally given due to potential risk of bleeding, particulife of 4 h. This short half-life makes the efficacy of ondanse- larly for surgical staple lines. tron for longer-lasting prophylaxis, such as in the ambulatory Patient-controlled analgesia (PCA) was developed to surgery setting, questionable. The incidence of post-­ allow intravenous administration of analgesics in an incredischarge nausea and vomiting (PDNV) after ambulatory mental fashion, so that respiratory depression and heavy surgery is higher than PONV and has been reported to be as sedation could be avoided. PCA use is highly recommended high as 50% in patients who did not experience for the severely obese patient, compared to intermittent PONV. Laparoscopic sleeve gastrectomy, currently the most bolus dosing [43]. Bupivacaine infusion devices for concommonly performed bariatric procedure, is particularly tinuous postoperative infiltration of the surgical wound emetogenic. One Polish study demonstrated an 8.2% inci- have been developed to avoid the risk of respiratory depresdence for LSG, compared to 1.4% for laparoscopic Roux-­ sion associated with opioids. Mixed results have been seen en-­ Y gastric bypass, with the use of enhanced recovery for potential benefits of heating and humidifying carbon pathways [39]. Aggressive antiemetic protocols using multi- dioxide (CO2) insufflation for postoperative pain relief in ple agents, and sparing opioids as able, seem to be the key to laparoscopic surgery. The European Association for reducing this rate. Endoscopic Surgery practice guidelines state that “the clinBesides adequate hydration with intravenous fluids, anti- ical benefits of warmed, humidified insufflation gas are emetic strategies covering a longer duration should be minor and contradictory” [44]. Theoretically, intraperitoemployed [40]. Multiple agents with different mechanisms neal (IP) administration of local anesthetics can provide of action (multimodal therapy) are more effective than a sin- analgesia without opioid-­related complications. A single gle agent. Dexamethasone, 8 mg, intravenously administered randomized clinical trial examined the use of continuous IP at the beginning of surgery combined with the transdermal infusion in laparoscopic adjustable gastric banding [45]. A cholinergic antagonist scopolamine is an effective longer-­ statistically significant decrease in VAS was noted in the IP lasting strategy. For patients with several risk factors, addi- infusion group with no differences in shoulder pain and tional medications such as haloperidol (0.5–1 mg) or additional medication use. promethazine (12.5 mg) can be added. Both can result in Obese patients with obstructive sleep apnea syndrome oversedation and should be administered with caution in (OSAS) appear to be more sensitized to sedation than normal patients with significant OSA. For patients with a history of individuals. Mortality may occur in OSAS patients after intractable nausea and vomiting, the neurokinin-1 receptor minimal doses of anesthetics or sedatives due to a change in antagonist aprepitant, 40 mg p.o., administered before sur- airway tone resulting in obstruction [46]. gery could be added and continued in the postoperative The ASA Guideline for Patients with Obstructive Sleep period. Opioid-sparing analgesia, beginning in the preopera- Apnea Syndrome (OSAS) is intended to improve perioperative period and continued throughout the intraoperative tive care and reduce the risk of adverse outcomes in patients period, and utilization of total intravenous anesthesia (TIVA) with OSAS who receive sedation, analgesia, or anesthesia. can help to decrease the rate of PONV in these patients [41, During preoperative evaluation, the severity of the patient’s 42]. Non-pharmacological methods such as acupuncture and OSAS, the type of surgery and anesthesia, and the requireacupressure have limited efficacy, but may be worthwhile ment for postoperative opioid analgesics should be considadjuncts. ered [37]. There is tremendous interest in establishing enhanced recovery after surgery (ERAS) guidelines that utilize opioid-sparing strategies for analgesia [42, 47], includPostoperative Analgesia ing the use of preoperative acetaminophen and gabapentinoids, and intraoperative dexmedetomidine, transAdequate postoperative pain relief is critical for patient com- versus abdominis plane or rectus sheath blocks [48], ketfort, pulmonary toilet, and early ambulation. Postoperative amine, esmolol infusion and lidocaine infusion with the analgesia must be balanced to avoid opioid-related compli- goals of decreased opioid-related adverse events, rates of cations like PONV, sedation, ileus, and respiratory depres- PONV [49], and shortened time to discharge or even same-­ 100 day laparoscopic sleeve gastrectomy [50]. However, there is significant heterogeneity among published reports of ERAS [51], and further work is needed in this area. Question Section 1. The obese patient is at increased risk for respiratory complications due to: A. Hypoxemia B. Higher respiratory rates C. Lower tidal volumes D. Decreased functional residual capacity E. Obstructive sleep apnea F. All of the above 2. The “sniffing” position of an obese patient during anesthesia induction is preferred to the supine position. A. True B. False 3. At the end of a laparoscopic sleeve gastrectomy, a 64-inchtall, 120-kg anesthetized patient is noted to have zero train-offour twitch and 1 post-tetanic count. The rocuronium-induced neuromuscular blockade should be reversed with: A. Neostigmine 0.08 mg/kg with glycopyrrolate B. Sugammadex 2 mg/kg at ideal body weight C. Sugammadex 4 mg/kg at total body weight D. Sugammadex 4 mg/kg at ideal body weight 4. A 60-inch-tall, 100-kg woman presents for bariatric surgery. Any of the following dosing would be appropriate except: A. Succinylcholine 50 mg for intubation B. Rocuronium 54 mg for intubation C. Propofol 140 mg for induction D. Succinylcholine 100 kg for intubation References 1. Littleton SW. Impact of obesity on respiratory function. Respirology. 2012;17(1):43–9.. [Review] 2. Masoomi H, Reavis KM, Smith BR, Kim H, Stamos MJ, Nguyen NT. Risk factors for acute respiratory failure in bariatric surgery: data from the nationwide inpatient sample, 2006-2008. Surg Obes Relat Dis. 2013;9:277–81. 3. Pouwels S, Smeenk FW, Manschot L, Lascaris B, Nienhuijs S, Bouwman RA, Buise MP. Perioperative respiratory care in obese patients undergoing bariatric surgery: implications for clinical practice. Respir Med. 2016;117:73–80. 4. Schumann R, Shikora SA, Sigl JC, Kelley SD. Association of metabolic syndrome and surgical factors with pulmonary adverse events, and longitudinal mortality in bariatric surgery. Br J Anaesth. 2015;114:83–90. 5. Glance LG, Wissler R, Mukamel DB, Li Y, Diachun CA, Salloum R, et al. Perioperative outcomes among patients with the modified metabolic syndrome who are undergoing noncardiac surgery. Anesthesiology. 2010;113(4):859–72. H. J. M. Lemmens et al. 6. Jones RL, Nzekwu MM. The effects of body mass index on lung volumes. Chest. 2006;130(3):827–33. 7. Pelosi P, Croci M, Ravagnan I, Tredici S, Pedoto A, Lissoni A, et al. The effects of body mass on lung volumes, respiratory mechanics, and gas exchange during general anesthesia. Anesth Analg. 1998;87(3):654–60. 8. Lecube A, Sampol G, Lloberes P, Romero O, Mesa J, Morell F, et al. Asymptomatic sleep-disordered breathing in premenopausal women awaiting bariatric surgery. Obes Surg. 2010;20(4):454–61. 9. Mokhlesi B, Tulaimat A, Faibussowitsch I, Wang Y, Evans AT. Obesity hypoventilation syndrome: prevalence and predictors in patients with obstructive sleep apnea. Sleep Breath. 2007;11(2):117–24. 10. Jense HG, Dubin SA, Silverstein PI, O’Leary-Escolas U. Effect of obesity on safe duration of apnea in anesthetized humans. Anesth Analg. 1991;72(1):89–93. 11. Dixon BJ, Dixon JB, Carden JR, Burn AJ, Schachter LM, Playfair JM, et al. Preoxygenation is more effective in the 25 degrees head­up position than in the supine position in severely obese patients: a randomized controlled study. Anesthesiology. 2005;102(6):1110– 5.. discussion 1115A 12. Heard A, Toner AJ, Evans JR, Aranda Palacios AM, Lauer S. Apneic oxygenation during prolonged laryngoscopy in obese patients: a randomized, controlled trial of buccal RAE tube oxygen administration. Anesth Analg. 2017;124(4):1162–7. 13. Gander S, Frascarolo P, Suter M, Spahn DR, Magnusson L. Positive end-expiratory pressure during induction of general anesthesia increases duration of nonhypoxic apnea in morbidly obese patients. Anesth Analg. 2005;100(2):580–4. 14. Carron M, Zarantonello F, Tellaroli P, Ori C. Perioperative noninvasive ventilation in obese patients: a qualitative review and meta-­ analysis. Surg Obes Relat Dis. 2016;12(3):681–91. 15. Bohm SH, Thamm OC, von Sandersleben A, Bangert K, Langwieler TE, Tusman G, et al. Alveolar recruitment strategy and high positive end-expiratory pressure levels do not affect hemodynamics in morbidly obese intravascular volume-loaded patients. Anesth Analg. 2009;109(1):160–3. 16. Lemmens HJ, Bernstein DP, Brodsky JB. Estimating blood volume in obese and morbidly obese patients. Obes Surg. 2006;16(6):773–6. 17. Janmahasatian S, Duffull SB, Ash S, Ward LC, Byrne NM, Green B. Quantification of lean bodyweight. Clin Pharmacokinet. 2005;44:1051–65. 18. Dong D, Peng X, Liu J, Qian H, Li J, Wu B. Morbid obesity alters both pharmacokinetics and pharmacodynamics of propofol: dosing recommendation for anesthesia induction. Drug Metab Dispos. 2016 Oct;44(10):1579–83. 19. Blaudszun G, Lysakowski C, Elia N, Tramer MR. Effect of perioperative systemic alpha2 agonists on postoperative morphine consumption and pain intensity: systematic review and meta-analysis of randomized controlled trials. Anesthesiology. 2012;116: 1312–22. 20. Hussain Z, Curtain C, Mirkazemi C, Zaidi STR. Peri-operative medication dosing in adult obese elective surgical patients: a systematic review of clinical studies. Clin Drug Investig. 2018;38(8):673–93. 21. Gupta K, Prasad A, Nagappa M, Wong J, Abrahamyan L, Chung FF. Risk factors for opioid-induced respiratory depression and failure to rescue: a review. Curr Opin Anaesthesiol. 2018;31(1):110–9. 22. Vesely A, Fisher JA, Sasano N, Preiss D, Somogyi R, El-Beheiry H, Prabhu A, Sasano H. Isocapnic hyperpnoea accelerates recovery from isoflurane anaesthesia. Br J Anaesth. 2003;91:787–92. 23. Liu FL, Cherng YG, Chen SY, Su YH, Huang SY, Lo PH, Lee YY, Tam KW. Postoperative recovery after anesthesia in morbidly obese patients: a systematic review and meta-analysis of randomized controlled trials. Can J Anaesth. 2015;62(8):907–17. 24. Singh PM, Borle A, McGavin J, Trikha A, Sinha A. Comparison of the recovery profile between desflurane and sevoflurane in patients 8 Anesthetic Considerations undergoing bariatric surgery-a meta-analysis of randomized controlled trials. Obes Surg. 2017;27(11):3031–9. 25. Thilen SR, Hansen BE, Ramaiah R, Kent CD, Treggiari MM, Bhananker SM. Intraoperative neuromuscular monitoring site and residual paralysis. Anesthesiology. 2012;117(5):964–72. 26. Murphy GS, Szokol JW, Avram MJ, Greenberg SB, Marymont JH, Vender JS, Gray J, Landry E, Gupta DK. Intraoperative acceleromyography monitoring reduces symptoms of muscle weakness and improves quality of recovery in the early postoperative period. Anesthesiology. 2011;115(5):946–54. 27. Loupec T, Frasca D, Rousseau N, Faure JP, Mimoz O, Debaene B. Appropriate dosing of sugammadex to reverse deep rocuronium-­ induced neuromuscular blockade in morbidly obese patients. Anaesthesia. 2016;71(3):265–72. 28. Schumann R, Jones SB, Ortiz VE, Connor K, Pulai I, Ozawa ET, et al. Best practice recommendations for anesthetic perioperative care and pain management in weight loss surgery. Obes Res. 2005;13(2):254–66. 29. American Society of Anesthesiologists Task Force on Central Venous Access, Rupp SM, Apfelbaum JL, Blitt C, Caplan RA, Connis RT, Domino KB, Fleisher LA, et al. Practice guidelines for central venous access: a report by the American society of anesthesiologists task force on central venous access. Anesthesiology. 2012;116(3):539–73. 30. Anesthesia Closed Claims Project. http://depts.washington.edu/ asaccp/projects. Last accessed 15 Apr 2019. 31. Pham Q, Lentner M, Hu A. Soft palate injuries during orotracheal intubation with the videolaryngoscope. Ann Otol Rhinol Laryngol. 2017;126(2):132–7. 32. Savoldelli GL, Schiffer E, Abegg C, Baeriswyl V, Clergue F, Waeber JL. Learning curves of the Glidescope, the McGrath and the Airtraq laryngoscopes: a manikin study. Eur J Anaesthesiol. 2009;26(7):554–8. 33. Stankiewicz-Rudnicki M, Gaszynski W, Gaszynski T. Assessment of ventilation distribution during laparoscopic bariatric surgery: an electrical impedance tomography study. Biomed Res Int. 2016;2016:7423162. 34. Gupta R, Gan TJ. Peri-operative fluid management to enhance recovery. Anaesthesia. 2016;71(Suppl 1):40–5. 35. Srinivasa S, Taylor MH, Singh PP, Yu TC, Soop M, Hill AG. Randomized clinical trial of goal-directed fluid therapy within an enhanced recovery protocol for elective colectomy. Br J Surg. 2013;100:66–74. 36. Weingarten TN, Kendrick ML, Swain JM, Liedl LM, Johnson CP, Schroeder DR, et al. Effects of CPAP on gastric pouch pressure after bariatric surgery. Obes Surg. 2011;21(12):1900–5. 37. Gross JB, Bachenberg KL, Benumof JL, Caplan RA, Connis RT, Cote CJ, et al. Practice guidelines for the perioperative management of patients with obstructive sleep apnea: a report by the American Society of Anesthesiologists task force on perioperative management of patients with obstructive sleep apnea. Anesthesiology. 2006;104(5):1081–93.. quiz 1117–1088 101 38. Kurrek MM, Cobourn C, Wojtasik Z, Kiss A, Dain SL. Morbidity in patients with or at high risk for obstructive sleep apnea after ambulatory laparoscopic gastric banding. Obes Surg. 2011;21(10): 1494–8. 39. Major P, Stefura T, Małczak P, et al. Postoperative care and functional recovery after laparoscopic sleeve gastrectomy vs. laparoscopic roux-en-Y gastric bypass among patients under ERAS protocol. Obes Surg. 2018;28:1031–9. 40. Schuster R, Alami RS, Curet MJ, Paulraj N, Morton JM, Brodsky JB, et al. Intra-operative fluid volume influences postoperative nausea and vomiting after laparoscopic gastric bypass surgery. Obes Surg. 2006;16(7):848–51. 41. Ziemann-Gimmel P, Goldfarb AA, Koppman J, Marema RT. Opioid-free total intravenous anaesthesia reduces postoperative nausea and vomiting in bariatric surgery beyond triple prophylaxis. Br J Anaesth. 2014;112(5):906–11. 42. Andersen LP, Werner MU, Rosenberg J, Gögenur I. Analgesic treatment in laparoscopic gastric bypass surgery: a systematic review of randomized trials. Obes Surg. 2014;24(3):462–70. 43. Graves DA, Batenhorst RL, Bennett RL, Wettstein JG, Griffen WO, Wright BD, et al. Morphine requirements using patient-controlled analgesia: influence of diurnal variation and morbid obesity. Clin Pharm. 1983;2(1):49–53. 44. Neudecker J, Sauerland S, Neugebauer E, Bergamaschi R, Bonjer HJ, Cuschieri A, et al. The European association for endoscopic surgery clinical practice guideline on the pneumoperitoneum for laparoscopic surgery. Surg Endosc. 2002;16(7):1121–43. 45. Sherwinter DA, Ghaznavi AM, Spinner D, Savel RH, Macura JM, Adler H. Continuous infusion of intraperitoneal bupivacaine after laparoscopic surgery: a randomized controlled trial. Obes Surg. 2008;18(12):1581–6. 46. Young T, Peppard PE, Gottlieb DJ. Epidemiology of obstructive sleep apnea: a population health perspective. Am J Respir Crit Care Med. 2002;165(9):1217–39. 47. Alvarez A, Goudra BG, Singh PM. Enhanced recovery after bariatric surgery. Curr Opin Anaesthesiol. 2017;30(1):133–9. 48. Said AM, Balamoun HA. Continuous transversus abdominis plane blocks via laparoscopically placed catheters for bariatric surgery. Obes Surg. 2017;27(10):2575–82. 49. Ng JJ, Leong WQ, Tan CS, Poon KH, Lomanto D, So JBY, Shabbir A. A multimodal analgesic protocol reduces opioid-related adverse events and improves patient outcomes in laparoscopic sleeve gastrectomy. Obes Surg. 2017;27(12):3075–81. 50. Badaoui R, Alami Chentoufi Y, Hchikat A, Rebibo L, Popov I, Dhahri A, Antoun G, Regimbeau JM, Lorne E, Dupont H. Outpatient laparoscopic sleeve gastrectomy: first 100 cases. J Clin Anesth. 2016;34:85–90. 51. Grant MC, Gibbons MM, Ko CY, Wick EC, Cannesson M, Scott MJ, McEvoy MD, King AB, Wu CL. Evidence review conducted for the Agency for Healthcare Research and Quality safety program for improving surgical care and recovery: focus on anesthesiology for bariatric surgery. Anesth Analg. 2018;14. 9 Components of a Metabolic and Bariatric Surgery Center Wayne J. English, D. Brandon Williams, and Aaron Bolduc Chapter Objectives After reading this chapter, the reader will have a better understanding of the: 1. Essential components necessary to operate a successful, high-quality metabolic and bariatric surgery center 2. Requirements to achieve national accreditation 3. Most common deficiencies encountered during an accreditation site survey Introduction The incidence of morbid obesity is increasing in epidemic proportions [1]. This alarming trend is being accentuated by frequent media-driven highlights and periodic calls for action from government officials. General surgeons are coming under increasing market pressures to provide surgical solutions for patients who seek significant and durable weight loss. Patients with class II and III obesity often carry multiple diagnoses, and many surgeons who had embarked on this specialty rapidly realized that the safest and most effective approach to managing these patients successfully is through a comprehensive management program [2]. There has been a 44.31% increase in the number of metabolic and bariatric procedures performed in the United States from 2011 through 2017 [3]. As the number of procedures being performed annually continues to increase, there is an even greater need for performing metabolic and bariatric sur- W. J. English (*) · D. B. Williams Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA e-mail: wayne.english@vumc.org A. Bolduc Department of Surgery, Augusta University Medical Center, Augusta, GA, USA gery with rigorous organizational and operational standards to ensure patients receive safe care. In this chapter, we will describe the essential components of a successful, comprehensive bariatric center including essential staff, ancillary personnel, material infrastructure, and educational and patient support strategies based on the Metabolic and Bariatric Surgery Accreditation and Quality Improvement Program (MBSAQIP) manual “Optimal Resources for Metabolic and Bariatric Surgery: 2019 Standards.” We will also briefly describe some tips to prepare for an MBSAQIP site survey and outline the most common deficiencies encountered during site surveys. he Metabolic and Bariatric Surgery T Accreditation and Quality Improvement Program In 2012, the American Society for Metabolic and Bariatric Surgery (ASMBS) and the American College of Surgeons (ACS) jointly developed the MBSAQIP standards to improve quality and facilitate access to care for patients. The decision to recommend surgery for obese patients requires multidisciplinary input to evaluate the indications for operation and to define and manage comorbidities properly. Institutions performing metabolic and bariatric surgery must have absolute commitment, organization, leadership, human resources, and physical resources to provide optimal care. Surgeons must demonstrate the necessary training, skills, and experience. Furthermore, high-quality surgical care requires data collection with reliable risk-adjusted measurements of outcomes. For metabolic and bariatric surgery centers to improve, surgeons require feedback, which is dependent on accurate data collection [4]. MBSAQIP semiannual data reports provide feedback for surgeons and their centers to help improve quality of care. Capturing uniform data elements for all participating MBSAQIP centers allows equal comparison of centers, which provides an opportunity for centers to continuously evolve and improve. © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_9 103 104 W. J. English et al. Metabolic and bariatric surgery procedure estimates in Periodic interrogation of the non-risk-­adjusted data provided 2016 demonstrate that approximately 92% of centers per- by MBSAQIP allows an intelligent and critical analysis of forming metabolic and bariatric surgery are MBSAQIP-­ patient outcomes, thereby indicating center components that accredited centers [5]. Ideally, all metabolic and bariatric may require special attention, and will allow centers to surgery centers in the United States will participate in the develop individualized quality improvement plans. MBSAQIP and assemble the essential components at the center to meet the required standards to achieve nationally recognized accreditation. MBS Committee ssential Components of a Metabolic E and Bariatric Surgery (MBS) Center The essential components of an MBS center include commitment to quality care, data collection with continuous quality improvement efforts, and designated leadership with the appropriate committee structure, surgical experience, critical care support, appropriate equipment and instruments, qualified call coverage, continuity of care pathways including long-term follow-up care and support groups structure. Leadership at the surgeon, integrated health and administrative levels are essential in developing a high-quality bariatric surgery center. A surgeon and integrated health partner would not be expected to completely understand the nuances of running the business aspect of a practice, nor should an administrator be expected to completely understand the clinical and technical aspects of surgery. Once the center has established a firm commitment for administrative support to develop and maintain a program, it is essential to assemble the “Leadership Trifecta,” a dedicated team comprised of the MBS Director (the leader of this elite group), MBS Coordinator, and Clinical Reviewer. The center should then reference the MBSAQIP standards as a guide to achieve their goals of developing and maintaining a high-quality MBS center. ata Collection and the Bariatric Surgery D Database An important requirement for any bariatric center is the ability to maintain a repository of information that includes patient demographics, comorbid factors, operative characteristics, and follow-up. Such a database will prove to be an invaluable resource for both clinical and research purposes. Ideally, patients will be followed for life, and there will occasionally be the need for rapid access to patient information. By collating and sorting data, a well-maintained database will demonstrate past performance and help direct future trends. For example, MBSAQIP focuses on perioperative safety and continuous quality improvement for all bariatric surgery centers. MBSAQIP provides semiannual risk-­adjusted reports (SAR) to determine how a center compares to other centers. The structure of a metabolic and bariatric center must consist of an MBS Committee that involves, at a minimum, the MBS Director, all surgeons performing metabolic and bariatric surgery at the center, the MBS Coordinator, the MBS Clinical Reviewer, and institutional administration representatives involved in the care of metabolic and bariatric surgical patients. The center must have a clearly defined mission, vision, goals, and objectives, which are discussed, and agreed upon, within the MBS Committee. The MBS Committee provides a setting for sharing best practices, reducing practice variation, responding to adverse events, and fostering a culture to improve patient care. Balanced representation is essential, and all surgical practices performing bariatric surgery at the center must participate in a collaborative manner focused on improving quality of care. The responsibility is upon the center, the metabolic and bariatric surgeon and, ultimately, the MBS Committee and MBS Director, to appropriately select patients and develop selection guidelines for the center relative to the center’s available resources and experience. The MBS Director and other team members of the MBS Committee at each center must embrace a culture of camaraderie and collaboration in order to report and analyze data, identify gaps of care, and implement strategies resulting in continuous quality improvement. Such efforts must be readily identified through the center’s MBS Committee. Major quality improvement projects that can result in decreasing rates of readmissions, reoperations, emergency department visits, length of stays, surgical site infections, leaks, or venous thromboembolism are extremely important in achieving safety and effectiveness of the procedures performed at a high-quality metabolic and bariatric surgery center. However, equally as important is the examination of clinical pathways of care to maximize the patient experience and improving overall patient satisfaction. The MBS Committee can also be considered a training ground for emerging and future leaders to refine their skills and abilities. Preparation for the center’s initial or the triennial site survey requires thorough documentation, within the meeting minutes, of the three meetings required, including at least one comprehensive review meeting annually. The comprehensive review meeting must be attended by all surgeons and proceduralists at the center. 9 Components of a Metabolic and Bariatric Surgery Center The center is required to review the two semiannual reports (SAR). For the third required meeting, the center is required to review the raw data available from the MBSAQIP data registry. Documentation of the review can be listed in generic terms and does not require patient-specific details. MBS Director The MBS Director, in collaboration with the MBS Committee, and the administration of the institution, organizes, integrates, and leads all metabolic and bariatric surgery-­related services throughout the accredited center. The Director is responsible for standardizing and integrating metabolic and bariatric patient care throughout the center and maintaining continuous compliance with accreditation standards. Continuous review of the standards will allow centers to identify, if present, any areas where there are gaps in compliance. All relevant staff must be educated on patient safety and complication recognition. An important goal would be to prevent “failure to rescue” situations, in which differences in mortality are proposed to result from the failure to timely recognize, and effectively manage, a postoperative complication. Additional training for the surgical teams and the integrated health personnel in postoperative complication recognition and management may improve outcomes [6]. The MBS Director is responsible for supervising all surgeons and proceduralists performing metabolic and bariatric procedures at the center and is obligated to report any significant ethical and/or quality deviations by surgeons and proceduralists to the appropriate institutional entities (e.g., chief of surgery, credentialing committee, medical staff, risk management, etc.). The MBS Director should have the authority to recommend plans for remediation or make formal recommendations to limit or redact privileges to the appropriate administrative or credentialing body at the center. MBS Coordinator Metabolic and bariatric surgery centers must have a designated MBS Coordinator who reports to and assists the MBS Director. The MBS Coordinator assists in center development, managing the accreditation process and ensuring continuous compliance with accreditation bodies, maintaining relevant policies and procedures, patient education, outcomes data collection, quality improvement efforts, and education of relevant institution staff in the various aspects of the metabolic and bariatric surgery patient with a focus on patient safety. It is essential for the MBS Coordinator to coordinate all services involved with the care of a metabolic and bariatric 105 patient. It is recommended that representatives for these services meet on a regularly scheduled manner to maintain compliance with the standards and address any issues that may arise with patient care. Services represented at these meetings could include, but are not limited to, designated bariatric floor, emergency department, radiology, lab, central supply, supply chain, bed control, and patient transportation. The MBS Coordinator supports the development of written protocols and education of nurses detailing the rapid communication and basic response to critical vital signs that is specifically required to minimize delays in the diagnosis and treatment of serious adverse events. One of the most important functions of the MBS Coordinator is to ensure all practicing surgeons and proceduralists are actively involved with the MBS Committee activity and process and quality improvement efforts. This can become very complicated if multiple surgical practices are involved at the center. The MBS Coordinator serves as the liaison between the institution and all surgeons performing metabolic and bariatric surgery at the center and, if applicable, all general surgeons providing call coverage. The MBS Coordinator will assist in coordinating and maintaining documentation of the call schedule provided by all covering surgeons. The MBS Coordinator must also work closely with the MBS Clinical Reviewer to assure timely submission of outcomes data. MBS Clinical Reviewer Managing data is a critical component of optimizing performance and quality of care. The center must designate a person, or department, that is accountable for gathering and entering data in a timely and accurate manner and providing outcomes reports when necessary. In an effort to eliminate any data collection bias, the designated MBS Clinical Reviewer should not be actively participating in a metabolic and bariatric surgery patient’s care (e.g., a surgeon, physician assistant, or advanced practice nurse). The MBS Clinical Reviewer will also fulfill ongoing training and recertification requirements, retrieve and enter long-term follow-up data on a compounding number of patients over time, and fulfill requests for patient data and reports to the MBS Coordinator and hospital quality improvement staff for analysis. The MBS Clinical Reviewer should work closely with the institution and clinicians to ensure that appropriate short-term and long-term data points are available in the medical records. It is critical that there is continuous communication between MBSCR and all surgeons performing MBS at the 106 center. It is imperative that the MBSCR has access to records from surgeons whose records are not integrated with the hospital electronic medical record system. This typically involves surgeons located off of the hospital grounds. A common site survey deficiency involves surgeons not allowing access to the records. Forms filled out by the surgeon or office staff containing specific data elements that can be biased are not considered appropriate for entry into the MBSAQIP data registry. The MBSCR must have full access to the records to ensure uniformity of the data collected in an unbiased manner. The MBSCR can contact the patient to collect data elements but cannot participate in direct patient care. Off-site third-party MBSCR services are available that can prepare your meeting agendas, reports, and meeting minutes. Healthcare Facility Accreditation Simply stated, hospitals pursue accreditation because it is required to receive payment from federally funded Medicare and Medicaid programs. However, there is consistent evidence that shows that accreditation programs improve the process of care provided by healthcare services and improve clinical outcomes of a wide spectrum of clinical conditions [7]. The accreditation process has the potential to significantly influence quality through a series of three mechanisms: coherence, organizational buy-in, and collective quality improvement action [8]. Healthcare facility accreditation ensures that the facility performed a comprehensive assessment of processes, policies, and procedures to provide safe care for all patients, including metabolic and bariatric surgery patients. The facility must be licensed by state or nationally recognized licensing authorities, if required by state law. Examples of licensing agencies include, but are not limited to, the Joint Commission, state health department, Det Norske Veritas, the American Osteopathic Association, the American Association for Accreditation of Ambulatory Surgery Facilities (AAAASF), the Accreditation Association for Ambulatory Health Care (AAAHC), or the Institute for Medical Quality (IMQ). Metabolic and Bariatric Surgeon “Verification” It is important that centers have at least one surgeon who dedicates a significant portion of their practice to metabolic and bariatric surgery. MBSAQIP requires that a center must have at least one “Verified” surgeon performing metabolic and bariatric surgery. Surgeon “Verification” recognizes the specialized skills of the dedicated metabolic and bariatric W. J. English et al. surgeon and is based on lifetime procedure volume of over 100 stapling procedures and a volume of at least 25 stapling procedures annually. It is essential for the “Verified” surgeon to document 24 h of bariatric-specific CME credits per triennial cycle. CME credits will not count unless the certificate specifically states the activity was bariatric-specific. Generic certificates without CME credit itemization will not apply to the total number of credits required for “Verification.” In addition to attending conferences related to obesity medicine and surgery to fulfill this requirement, there are a number of online opportunities to meet compliance with the “Verified” CME requirements through journal article reviews (Surgery for Obesity and Related Diseases, Journal of the American College of Surgeons), societies (ASMBS, The Obesity Society, Obesity Medicine Association), and the American Board of Obesity Medicine. I nstitutional Requirements for Metabolic and Bariatric Surgeon Credentialing Well-trained surgeons are vital in delivering high-quality care to metabolic and bariatric surgery patients in a consistent manner. Centers performing metabolic and bariatric surgery should be committed to improving outcomes and require surgeons to obtain the skills necessary to achieve optimal outcomes. The institution’s credentialing body should follow nationally recognized credentialing guideline regarding surgeon experience and training for surgeons seeking metabolic and bariatric surgery privileges which are separate from general surgery guidelines. Credentialing guidelines, such as that collectively produced by the joint task force involving the ASMBS, the Society of American Gastrointestinal and Endoscopic Surgeons (SAGES), ACS, and the Society for Surgery of the Alimentary Tract (SSAT), aim to ensure that bariatric surgeons have undergone appropriate training and have achieved a certain minimum level of skill to safely perform bariatric surgery and to recognize and treat complications [9–12]. ualified Metabolic and Bariatric Surgery Call Q Coverage All surgeons performing metabolic and bariatric surgery at a center must have qualified coverage at all times by a colleague who is responsible for the emergency care of a metabolic and bariatric surgery patient. Call coverage may involve one or more general surgeons who are not privileged to perform metabolic and bariatric surgery. The covering general surgeon must have general surgery privileges at the facility and must have completed formal education and training 9 Components of a Metabolic and Bariatric Surgery Center regarding a basic understanding of the (1) metabolic and bariatric procedures commonly performed at the center, (2) signs and symptoms of postoperative complications, and (3) management and care of the patient by a review of the center’s clinical pathways and protocols. Centers may elect to require general surgeons to perform as a first assistant for a designated number of procedures to become better acquainted with anatomy and potential complications. MBSAQIP participating centers are required to document a care pathway for metabolic and bariatric surgery patients who have had surgery elsewhere. Examples to address this issue include having complete coverage by the center’s credentialed metabolic and bariatric surgeons or general surgeons covering unassigned patients with metabolic and bariatric surgeon being available when needed. Smaller or solo surgeon centers may opt to obtain locum tenens bariatric surgery coverage. esignated Area for Metabolic and Bariatric D Surgery Patients It is important to standardize care with personnel that routinely have contact with metabolic and bariatric surgery patients. The center must have a dedicated metabolic and bariatric surgery floor, or designated group of beds, maintained in a consistent area of the facility. Designated Personnel The center must involve an integrated health approach to the metabolic and bariatric surgery patient. The optimal care of the metabolic and bariatric surgery patient requires specialized training, education, and experience, such as that obtained with Certified Bariatric Nurse (CBN®) certification. It is essential that a consistent operating room team is assigned to optimize patient safety and the flow of the procedure. As needed, the center must provide access or referral to registered nurses, advanced practice nurses, or other physician extenders, registered dietitians, psychologist, psychiatrist, social worker, or other licensed behavioral healthcare provider, and physical/exercise therapists. egistered Dietitians and the Dietary Evaluation R Registered dietitians (RD) are essential for the success of any bariatric center and especially the success of the patient undergoing bariatric surgery. They provide nutritional counseling for patients preparing for surgery and assist patients with postoperative nutritional counseling and maintenance. Dietitians have been shown to improve outcomes in the postoperative bariatric surgery patient. A retrospective analysis of a prospective database was performed on bariatric surgery 107 patients who were followed up by either a surgeon alone or by a surgeon and RD for initial postoperative visit. Patients in the RD follow-up group had significantly fewer readmissions due to dietary-related problems and more favorable 3-month change in serum thiamine, high-density lipoprotein, and triglycerides. Additionally, patients trended toward a lower number of minor complications [13]. An additional important role for nutritional counselors is the assessment of a candidate’s ability to comply with the required dietary modifications imposed by bariatric surgery. Patients should have an appreciable degree of motivation to accept and reliably follow dietary guidelines and should have a minimum level of understanding of the potential repercussions of nonadherence. Dietitians are in an ideal position to gauge patients’ eligibility and compliance from a cognitive and motivational perspective [14, 15]. Of all the dietary macronutrients, proteins are the most valuable and essential. Patients’ dietary regimens must meet their daily requirements for these nutrients. Nutritional counselors are trained to deliver advice on the proper identification of protein sources and the preparation of foodstuffs to render them more palatable. They will also advise patients on proper chewing and swallowing techniques and give tips on how to avoid consuming liquids concomitantly with solids. Patients are required to know the hazards of insufficient fluid intake and are counseled to imbibe through the day, pausing before meals. The daily administration of vitamins and supplements is a critical requisite for postoperative patients, and it is incumbent on surgeons to ensure that patients are meeting their nutritional needs. Surgeons have been held liable for complications of vitamin deficiencies, particularly neurological manifestations that may result from inadequate intake. The provision of appropriate access to nutritional counselors and opportunities for periodic laboratory assessment of nutritional parameters may reduce the occurrence of these indefensible and undesirable outcomes. sychologist and the Psychological Evaluation P The primary objective for the psychosocial evaluation for patients with Class II and III severe obesity interested in undergoing metabolic and bariatric surgery is to screen for risk factors and identify potential challenges that may contribute to a poor postoperative outcome. Patients with Class II and III obesity often carry a diagnosis of depression, anxiety, and other stress-related conditions. There are often problems with body image and with low and demoralized self-esteem. Psychologists offer invaluable support in assessing patients’ mental conditions and counsel the patient to withstand the lifelong changes imposed by bariatric surgery. Several instruments for psychological assessment of morbidly obese patients in the preoperative and postoperative periods have been developed and validated and are in wide use. 108 Pre- and postoperative education of patients presenting for weight loss surgery is more critical as compared to that of patients preparing for non-bariatric surgery. For example, knowledge of the “rules of eating” and the “rules of vomiting” is essential for the positive outcome of gastric restrictive surgery. Discrepancies between patients’ weight goals, “ideal” or healthy weight for post-obese individuals and ­realistic weight loss based on body composition and energy balance, contribute to subjective assessment of quality of life after bariatric surgery. It is common for bariatric patients to experience postoperative nausea, depression, and possibly remorse for several months following surgery. Many patients who successfully lose weight may experience jealousy from close friends and relatives. Difficulty exists for the surgeon in delineating the physical from the psychological etiologies of many postoperative symptoms that afflict patients. Stress factors may significantly influence a patient’s ability to achieve maximum and durable benefit or may even predispose to deviations and weight regain. Preoperative education, evaluation, and preparation, although essential, will not identify nor eliminate all the potential problems. Psychological intervention is sometimes useful in achieving overall patient stability and emotional well-being, thereby underscoring its important role throughout the entire preoperative and postoperative course of a patient’s treatment experience [16, 17]. To further emphasize the importance of having continued behavioral health services available in the postoperative period, a review of patients undergoing bariatric surgery in Pennsylvania, patients with a diagnosed mental health disorder had 34% greater odds of 30-day readmission relative to patients with no diagnosed mental health disorder. Patients with major depressive disorder/bipolar disorder had 46% greater odds of 30-day readmission compared with patients with no major depressive disorder/bipolar disorder diagnosis [18]. Addiction transfer is an issue that should be monitored in the postoperative metabolic and bariatric surgery patient. It has been theorized that obesity-induced dysregulation of the dopamine reward processing can result in compensatory overeating, which may be reversed after RYGB. After surgery, patients may seek alternative means, such as alcohol and illicit substances, to obtain the dopaminergic reward previously obtained with food [19]. Many factors may significantly influence a patient’s ability to achieve maximum and durable benefit or may even predispose to deviations and weight regain. Preoperative education, evaluation, and preparation is essential, although it is likely that it will not identify, nor eliminate, all potential issues that may arise after surgery. A systematic review revealed an increased risk, with an odds ratio of 3.8, for self-harm and suicide attempt in patients undergoing bariatric surgery compared to matched control W. J. English et al. subjects. A study in Sweden looked at patients who underwent primary RYGB between 2001 and 2010 found that patients were nearly 2.85 times more likely to make a suicide attempt than the general population reference group. A number of psychosocial issues that can potentially be attributed to suicide must be considered in the metabolic and bariatric surgery patient, including lack of improvement in quality of life after surgery, continued or recurrent physical mobility restrictions, persistence or recurrence of sexual dysfunction and relationship problems, low self-esteem, a history of child maltreatment, inadequate weight loss, or weight regain [20–22]. Appropriate Equipment and Instruments For optimal safety and excellent outcomes, a successful bariatric surgery center requires the facility have appropriate furniture and equipment to accommodate patients within the weight limits established by the program. Patients with morbid obesity should feel welcome and safe upon entering any part of the center where they might receive care as an outpatient or inpatient (e.g., clinic, lab, endoscopy suite, radiology department, hospital admitting, and emergency department). At a minimum, they must be accommodated with appropriate weight capacity furniture, exam tables, wheelchairs, and toilet facilities in the waiting areas and examination rooms in all such areas. Often, wall-mounted toilets may not have a sufficient weight capacity. Installing a floor support is a simple solution to this problem (Fig. 9.1). The same considerations apply to weight scales and sphygmomanometer cuffs. All inpatient units of the hospital that the bariatric patient might encounter (e.g., pre-op holding, operating rooms, PACU, surgical ward, radiology, endoscopy, and ICU) must have all of the above in addition to appropriate clothing, sequential compression device sleeves, stretchers, operating room tables, hospital beds, shower rooms, doorways, walkers, and transfer equipment. Particular consideration must be given to having immediate access to equipment that may be rarely used, but can be indispensable in a crisis, such as extra-long surgical instruments (open and laparoscopic), difficult airway devices, and a lift for a patient who has collapsed in a chair or on the floor and cannot get up. Centers must have a thorough written system of documenting the weight capacity of all relevant equipment according to the manufacturer. Most importantly, this information must be easily accessible to all staff members caring for the bariatric patient. Centers may opt to have rental agreements for equipment that is not available on-site, but such agreements must have guaranteed delivery timeframes. Should a program choose to care for patients who exceed the weight capacity of any key equipment, such as the CT 9 Components of a Metabolic and Bariatric Surgery Center 109 Fig. 9.1 Examples of floor supports. Courtesy of Big John Products Inc. scanner or fluoroscopy tables, then there must be a documented care pathway for such patients with specific attention to the preoperative patient counseling and consent regarding this limitation and how it will be handled. Bariatric surgery centers must have a dedicated metabolic and bariatric surgery unit or designated group of beds maintained in a consistent area. There must be well established, properly managed, and ongoing in-service education programs for all the nurses and staff caring for the bariatric patients. The educational programs must ensure maintenance of competency in at least the following areas: 1. Recognizing the signs and symptoms of postoperative complications. All nurses and staff must be able to quickly recognize evidence of complications (e.g., pulmonary embolus, respiratory distress or hypoventilation, anastomotic leak, infection, or bowel obstruction) to facilitate timely intervention and help prevent failure to rescue. 2. Understanding the treatment of obesity as a disease, as endorsed by the American Medical Association [23]. Patients with obesity have likely long suffered from pejorative language and attitudes associated with the social stigma of obesity. Unfortunately, much of the contempt and criticism may have come from healthcare professionals [24]. Therefore, it is essential that the entire patient care team receive sensitivity training to ensure provision of compassionate and supportive care for bariatric patients. This training should emphasize the burden of obesity, its detrimental impact on health and quality of life, and include an appreciation for the necessary role of metabolic and bariatric surgery. 3. Safe patient transfer and mobilization. The staff should be well educated in fundamental transfer techniques, as well as where to obtain relevant equipment and how to properly use it to avoid injury to themselves or to bariatric patients when assisting with transfer or mobilization. Critical Care Support Bariatric centers, namely, the bariatric surgery committee and medical director, must develop patient selection protocols and guidelines relative to the available resources and experience level of the center. For example, patients at risk 110 for major organ failure should only be treated in centers with immediate access to all the medical resources that such patients might require. On occasion, however, metabolic and bariatric surgery patients will require unexpected critical care, and centers must ensure that patients receive appropriate care. At all times, centers must have immediate, on-site availability of personnel capable of administering advanced cardiac life support. Also, centers must have the ability to stabilize critically ill bariatric patients with resources such as difficult airway equipment, ventilator support, and hemodynamic monitors. If a facility is unable to manage on-site all aspects of critical care, namely, pulmonology/critical care, cardiology, and nephrology, the center must have the ability to transfer patients to a higher level of care. In this situation, the center must have a signed written transfer agreement that details the transfer plan of patients to other facilities capable of managing the full spectrum of complications. Anesthesiology for Bariatric Surgery Just as pediatric patients are not merely “little people,” patients with morbid obesity are not typical patients who happen to be very large. Failure to appreciate the sequelae of morbid obesity fully will result in unacceptable morbidity and mortality. Anesthesiologists, certified registered nurse anesthetists (CRNAs), and anesthesiologist assistants (AAs) who are charged with managing bariatric patients undergoing major surgery should be experienced in diagnosing and treating immediate or imminent life-threatening ­conditions, such as a difficult airway, hypoventilation syndrome, sleep apnea, congestive heart failure, renal insufficiency, and venous thromboembolism, to name a few. Anesthesiologists, CRNAs, and AAs who will manage bariatric patients undergoing laparoscopic operations must be cognizant of the pulmonary and hemodynamic changes that occur upon establishing and maintaining prolonged pneumoperitoneum. These cases are not to be taken lightly and certainly not to be delegated to inexperienced staff. There should, at all times, be more than one experienced anesthesiology staff member or qualified, similarly experienced, anesthesia nursing personnel available who can provide care in accordance with state laws governing their scope of ­practice [25]. The collaboration between experienced bariatric surgeons, anesthesiologists, and the OR staff represents a powerful intellectual combination that greatly benefits patients. Perioperative recommendations by “bariatric” anesthesiologists are invaluable in promoting favorable outcomes in patient safety and comfort. W. J. English et al. Continuum of Care Although bariatric surgery has been practiced for several decades, it remains an evolving discipline. Many biological features of the metabolic, endocrine, nutritional, and psychological manifestations of morbid obesity are just now being revealed. Patients undergo dramatic physical, metabolic, physiological, and psychological changes in many aspects, often by unclear mechanisms. There is, however, a constant accrual of voluminous information as more research is expended on this growing field. Understandably, morbidly obese patients considering weight loss surgery, or who are recovering from these operations, often possess insatiable appetites for information. Although all members of a bariatric surgery center should be reasonably abreast of established and emerging knowledge, it is incumbent on the physicians to assume the role of experts in the scientific aspects of bariatric medicine. Physicians, and their physician assistants and nurses, represent an invaluable reference source to patients. Their information is best delivered to patients in the way they learn best, both in one on one patient interaction and in a group setting where discussion can flow freely between inquisitive patients (and their family members) and bariatric center personnel. Informal group settings may provide the most efficient method of disseminating information. Two examples of effective group activities are the preoperative educational workshops and the support groups. he Preoperative Educational Workshop T Educational seminars, or workshops, represent the gateway into most metabolic and bariatric surgery centers. The primary purpose of these sessions is to educate the patient and gain critical information to ensure safety for the patient. Evidence shows that teaching about the specific risks and benefits and how each procedure may fit a different person has the effect of providing informed consent. Some patients will choose to change their procedure (15%), and some will choose not to undergo surgery (9%) [26]. This is a mutually beneficial exercise between prospective patients and bariatric center health providers. Patients gain acquaintance, in a broad sense, with the scheme of the center, the pathway they will follow in their preoperative evaluation, the inherent features that pertain to different laparoscopic and possibly open operations, the risks and benefits of each procedure, and the importance of adhering to feeding and exercise guidelines. Particular emphasis is placed on the need for lifelong follow-up and periodic assessment of nutritional parameters. The center’s health providers, on the other hand, may perform initial evaluations of the patients, review their nutritional and medical backgrounds, and consider patients’ eligibility for surgery. Patients who are deemed to require specialist work-ups are 9 Components of a Metabolic and Bariatric Surgery Center appropriately referred. The duration of these workshops varies from center to center but includes informative presentations by the center’s staff with audiovisual aids and printed material for future review. Family members should be encouraged to attend. Patients and their family members should be given ample opportunity to participate in a dedicated free-flowing discussion. Support Groups Support groups are designed for patients who are in the recovery phase of their operations. Although prospective patients are encouraged to attend, these sessions should be primarily targeted to addressing medical, nutritional, psychological, and social issues experienced by postoperative patients. Patients undergo dramatic changes that affect every sphere of life, some of which may be difficult to accept. Patients greatly benefit from the tips and advice delivered by more experienced patients and from the center staff. Additional gain may be achieved by holding informal educational lectures, in lay terms, by a variety of invited speakers. It is important that staff members of the bariatric center consistently attend support group sessions, moderating and monitoring the discussion to ensure that false information or misconceptions are not disseminated [27]. Currently, many centers have online support groups/blogs and use other forms of social media to keep in touch with patients and make them feel connected to the center. Following bariatric surgery, the inclusion of a support group as part of the treatment plan makes aftercare easier and more efficient for the patients, as well as for the physicians. The MBSAQIP accreditation requires that centers provide regularly scheduled organized and supervised support groups to metabolic and bariatric surgery patients. It is therefore obvious that institutions must ensure the availability of enough space to accommodate educational seminars and support group sessions. Moreover, such dedicated space should be furnished according to the needs of the bariatric patients and their families. Such a dedicated environment will clearly reassure patients that they are welcomed and will be a strong encouraging factor in optimizing their attendance and participation. Goals of Preoperative Patient Education and Teaching • Establishment of a process of informed consent that can be documented • Encouragement for compliance and praise • Education about life after surgery, including nutrition, exercise, and dieting techniques • Identification of problems • Identification and development of new kinds of self-nurturing 111 • Participation in a forum where others really “understand” the challenges and difficulties associated with “change,” even when the change is for the better • Creation of a friendly, safe atmosphere where patients can bring spouses, parents, and significant others so that they may also understand, encourage continuing success, and recognize their own personal issues related to major changes that they are experiencing with their loved one • Opportunity for curious potential patients in the community to come and learn from the “experts” in an environment of true caring and concern pecialty Consultants and Preoperative S Clearances Morbidly obese patients often have associated comorbid factors that negatively affect quality of life and often pose a significant risk on a day-to-day basis. Additionally, if unrecognized or inappropriately managed, these factors may be a cause of perioperative complications. Serious medical conditions associated with morbid obesity include cardiovascular, pulmonary, endocrine, metabolic, hematological, and many other diseases. The availability of consultants and experts in these fields is critical. Specialty physicians should be familiar with the particular pathophysiologic consequences of morbid obesity and should be able to ascertain with a certain degree of confidence the eligibility of candidates to withstand the rigors of major surgery and the required physical demands on patients in the postoperative period. Such consultants should be proficient in adequately preparing patients for general anesthesia, particularly regarding cardiac and pulmonary reserve, and the implementation of special preoperative patient respiratory training. All patients should undergo extensive evaluation prior to weight loss surgery. Careful initial history taking and clinical examinations will guide clinicians to diagnose previously unrecognized diseases. Risk factors associated with increased complication rates after surgery were identified in a number of published studies [28–31]. These risk factors include increasing age, male gender, increasing body mass index, mobility limitations, hypertension, prior history of a venous thromboembolism, coronary artery disease, myocardial infarction within the previous 6 months, angina, prior history of coronary intervention, congestive heart failure, history of stroke, bleeding disorder, smoking history, procedure type, procedure time greater than 3 h, obstructive sleep apnea, dyspnea, corticosteroid use, peripheral vascular disease, and liver disease. A center can consider utilizing one or more risk 112 prediction models established to assess the patient’s overall risk associated with metabolic and bariatric surgery. By following appropriate algorithms and considering particular risk factors, many known and undiagnosed conditions can be evaluated, not in an attempt to discourage or prevent operations, but with the goal of fully optimizing surgical outcomes by taking special perioperative precautions and additional supportive measures. W. J. English et al. As more patients participate in social networking platforms like Facebook, Twitter, and YouTube, there is an emerging trend in using social media within bariatric surgery centers. A University of Florida study suggested there might be a sixfold increase in the number of social media users in the next generation of physicians. The study found that only 13% of physician residents compared to 64% of medical students had Facebook profiles [32]. More healthcare professionals are using LinkedIn to keep in contact with other professionals as well. Sharing knowledge and informing Electronic and Remote Access patients using the social network can be of great benefit to your patients and for your center as it allows your profesto the Metabolic and Bariatric sional identity to develop. However, it is important to emphaSurgery Center size that if a center decides to integrate social media into The ability to obtain information instantly on any topic is the their practice, protecting the rights of patients and exhibiting results of the tremendous advancements made in the field of online professionalism are paramount. cyber technology, which includes access to the Internet, To guide physicians through the complex nature of social Telehealth, and social media. The positive benefits obtained media interaction with patients, the American College of through this achievement, however, present as a double-­ Physicians and the Federation of State Medical Boards pubedged sword. Internet and social media content is not regu- lished a position paper with recommendations about the lated or controlled. For the most part, metabolic and bariatric influence of social media on the patient–physician relationsurgery and obesity-related websites provide valuable infor- ship, the role of these media in public perception of physimation, but there are sites that do disseminate erroneous and cian behaviors, and strategies for physician–physician false information. communication that preserve confidentiality while best using For this reason, it is highly advisable to invest in creating these technologies (Table 9.1) [33]. electronic on-site resources that accurately reflects the mission and purpose of the bariatric surgery center. A website should contain, in lay terms, an explanation of the problems Metabolic and Bariatric Surgery of obesity and the available medical and surgical solutions. It in Adolescents should describe the physical and personal setup of the center, and the preoperative and postoperative patients will follow. The prevalence of childhood obesity and numerous obesity-­ Additionally, it would be of great benefit for the website to related comorbidities has risen exponentially over the past possess the ability to accept initial patient application forms several decades. In addition, a mounting body of scientific electronically, to be used as a basic screening tool prior to evidence demonstrating a high propensity for severely obese inviting the patient for the educational workshop. A chat adolescents to become severely obese adults has led to an room for patients and/or a social media platform where increase in the consideration and utilization of surgical patients could directly contact the surgery center personnel weight reduction procedures in this emerging population. may result in a modality that is more efficient and practical Adolescent bariatric surgery is safe and effective in treating than relying on conventional telephonic arrangements. patients with obesity. In addition to weight loss and comorEstablishing a HIPAA-compliant Telehealth network can bidity resolution, adolescents also benefit from prevention of allow better access to your center and may help improve many other conditions. The 2018 ASMBS pediatric metaoverall long-term follow-up with your patients. For many bolic and bariatric surgery guidelines recommend early rural bariatric centers, patients may need to travel many intervention to reduce progression of end-organ damage hours to attend preoperative seminars, educational sessions, [34]. This is further outlined and supported by data derived support groups, and routine postoperative visits. Patients can from Teen-Longitudinal Assessment of Bariatric Surgery simply travel to their local clinic or physician’s office, sign (Teen-LABS) and Treatment Options of Type 2 Diabetes in into the Telehealth network, and engage in a routine patient– Adolescents and Youth (TODAY) [35, 36]. physician encounter with their bariatric surgeon remotely, Metabolic and bariatric surgery should only be performed thus reducing some of the financial burden associated with on adolescent patients at an adolescent center accredited by high costs of fuel and lodging. Avoiding lengthy trips in the MBSAQIP. Essential personnel, trained in evaluating and dreaded weather conditions is an unequivocal benefit of caring for adolescent patients, must be available to ensure Telehealth as well, especially in the wintertime when road safe care for the adolescent patient and to optimize long-term conditions may be dangerous and not readily accessible. success after surgery. 9 Components of a Metabolic and Bariatric Surgery Center 113 Table 9.1 Activities of physicians online. The pros, the cons, and recommended conduct Online physician behavior Type of online activity Communicating with patients via email, text, and instant messaging Using social media to gather information about patients Referring patients to online educational resources and related information Physician blogging, microblogging, and physician postings of comments by others Physician posting of their own personal information on public social media sites Physician using electronic venues (e.g., text and Web) to discuss patient care with colleagues Pros Greater accessibility to patients and vice versa Cons Possible security breaches in confidentiality Can provide and receive immediate answers regarding nonurgent issues Absent or reduced face-to-face or telephone interactions Possible ambiguity or misinterpretation of information provided Sensitivity to source of information Allows observation of patients engaged in any risk-taking or health-averse behaviors Counsel patients or even intervene in an emergency Encourages patient empowerment through self-education Supplements resource-poor environments Allows advocacy and public health enhancement Introduces a physician “voice” into online medical conversations Can improve networking and communications Allows easy communication with colleagues Can create trust problems in the patient– physician relationship Patients may be unable to differentiate between good information sources and non-peer-reviewed materials that may provide inaccurate information Scam “patient” sites that misrepresent therapies and outcomes Allows for negative online comments that disparages patients and colleagues Can blur professional and personal boundaries Can have negative impact on how the individual and the profession are represented to the public Raises patient confidentiality concerns Possible unsecured networks and accessibility of what should be protected health information Recommended conduct Establish guidelines for the types of issues that are appropriate for online communication Reserve digital communication only for patients who maintain face-to-face follow-up Consider the intent of observing patients via their social media activities and how to address adverse findings Consider the impact on ongoing care Physician should review the information patients find in order to ensure the accuracy of the content Refer patients only to reputable websites and sources “Pause before posting” Consider the content of comments and the message it sends about a physician as an individual and the profession Physicians should maintain separate personas for their personal and professional online behavior Physicians should scrutinize what material they make available for public consumption Implement health information technology solutions for secure messaging and information sharing Follow institutional practice and policy for remote and mobile access of protected health information Adapted from Farnan et al. [33] Pediatric Medical Advisor (PMA) Every adolescent patient requires a pediatrician or equivalent provider who will participate in the preoperative and postoperative care of the adolescent patient and provide ongoing general pediatric medical oversight. In addition, the PMA can assist in the utilization of adolescent-specific s­ ubspecialty consultation when needed (i.e., sleep medicine, gastroenterology, endocrinology, hematology, nephrology, behavioral health, etc.). This individual should possess the necessary training and be credentialed in general pediatrics and/or pediatric subspecialty training (i.e., endocrinology, cardiology, gastroenterology, adolescent medicine). Responsibilities should also include assisting in the development of comorbid-specific treatment plans in conjunction with the patient’s primary care provider in order to optimize perioperative health. Adolescent Behavioral Specialist In order to achieve successful outcomes in adolescents, there must be assurances that patients and their families have the ability and the motivation to comply with recommended treatments preoperatively and postoperatively, including consistent use of recommended nutritional supplements. A history of reliable attendance at office visits for weight management and compliance with other medical needs is critical in determining compliance. 114 W. J. English et al. The adolescent patient and family must be able to demonstrate awareness of the general risks and benefits of metabolic and bariatric surgery as well as the dietary and physical activity requirements following a metabolic and bariatric procedure [37]. A psychologist, psychiatrist, or other qualified and independently licensed provider with specific training and credentialing in pediatric and adolescent care must perform this assessment. The practitioner must have experience in treating obesity and eating disorders as well as experience evaluating adolescent patients and families. The elements of behavioral assessment should include evidence for mature decision-making and awareness of potential risks and benefits of the proposed operation, documentation of the adolescent’s ability to provide surgical assent, evidence of appropriate family and social support mechanisms (i.e., engaged and supportive family members, caretakers, etc.), and clinically stable behavioral disorders (i.e., depression, anxiety, etc.) that have been satisfactorily treated. ommon Deficiencies Encountered During C the MBSAQIP Site Survey 1. Data entry of all procedures: In most cases, this involves incomplete entry of procedures due to having no MBSCR available or unavailability of medical records from surgeon office records. In some cases, deficiencies are cited because centers are performing investigational procedures without Institutional Review Board approval. Table 9.2 Top ten deficiencies encountered during a site survey The MBSAQIP accreditation process is laborious and involves triennial site visits by a surgeon surveyor. The site survey assesses whether the center is in compliance with the standards, including Case Volume, Commitment to Quality Care, Appropriate Equipment and Instruments, Critical Care Support, Continuum of Care, Data Collection, and Continuous Quality Improvement Processes. Additional requirements are necessary for stand-alone Children’s Hospitals and Ambulatory Surgery Centers. Each standard is comprised of various elements, and accreditation is only achieved if compliance is met with all components for each standard. Standard 6.1 7.1 6.2 % of sites with deficiency 5.98 5.01 4.85 Requirement Data entry of all procedures Institutional collaborative Data reports, quality metrics, and quality monitoring Process improvement initiatives Credentialing guidelines MBSCR Monitoring of safety culture Qualified call coverage Surgeon verification Facilities, equipment, and instruments 7.2 2.6 2.4 7.3 2.8 2.7 3.1 3.88 3.72 3.39 3.32 3.07 2.91 2.42 # of Deficiencies per Standard 140 120 100 80 60 40 20 0 123 75 67 9 DA R ST AN D DA R D 8 7 ST AN DA R D 6 D # of Deficiencies ST AN DA R ST AN D DA R 5 0 5 4 DA R D 3 D DA R ST AN D DA R ST AN 2 1 D ST AN 24 18 15 ST AN 7 DA R ST AN Fig. 9.2 Number of deficiencies per standard found on MBSAQIP site surveys In a review of 619 site surveys performed between September 2014 and August 2016, nearly one-quarter of MBSAQIP applicants had at least one deficiency. The Standards categories most often cited were related to quality and data, indicating opportunities for improvement (Fig. 9.2). 98.9% of centers with deficiencies were able to gain or maintain accreditation through corrective or remedial actions, demonstrating the effectiveness and potential developmental prospect of the accreditation process [38]. Understanding which standards are most commonly deficient following MBSAQIP site surveys allows centers to become better prepared for their accreditation or reaccreditation process. Table 9.2 describes the most common deficiencies found during site surveys. 9 Components of a Metabolic and Bariatric Surgery Center 115 2. Institutional collaborative: Many centers will review and effectively will strengthen accordingly. Because morbid their adverse events and its semiannual risk-adjusted obesity is a disease that globally affects patients, it can only report (SAR), but deficiencies are cited when there is no be managed by a diverse group of skilled individuals, all of documentation of such a review in the meeting whom are united in the common purpose of its eradication. minutes. The aim of surgical solutions should be permanent weight 3. Data reports, quality metrics, and quality monitoring: loss, and this will require the prolonged services of bariatriSimilar to #2, centers will typically review their out- cians, nutritionists, physical therapists, perhaps even spiritual comes, but deficiencies are cited when the center does healers, and marriage counselors. The fact is that there must not document in the MBS Committee meeting minutes be a comprehensive program in place, with the ability to evalthat this was reviewed at least three times annually. uate prospective patients in a comprehensive manner, and to 4. Process improvement initiatives: Most commonly, cen- prepare them mentally and physically for the rigors of surters will fail to document at least one quality, or process, gery and permanent lifestyle changes they will need to adopt. improvement project annually. Most importantly, defiThe most important element in the bariatric surgery cencient centers fail to address high outlier status on their ter is leadership from the surgeons and integrated health SARs. staff. Such a comprehensive center requires dedicated per5. Credentialing guidelines for metabolic and bariatric sonnel, a strong fundamental infrastructure, and total institusurgeons: Centers that are deficient in this standard most tional commitment. Most important of all, there needs to be commonly lack credentialing guidelines that are sepa- a pervasive philosophy that stretches across all institutional rate from general surgery credentialing guidelines. The departments and that accepts morbid obesity as a disease standards require centers to adhere to nationally recog- process, the management of which is a noble and dramatinized credentialing guidelines [9]. cally rewarding mission. 6. MBSCR: The most common reason for deficiencies in this standard is the MBSCR cannot enter data from all procedures and follow-up due to lack of access to office Question Section records from surgeon offices. All surgeons participating at the center must provide access to remain compliant 1. The Metabolic and Bariatric Committee must include all with this standard. of the following personnel except for: 7. Ongoing monitoring of safety culture: Centers that A. Surgeon Director who is actively performing metareceived a deficiency with this standard failed to review bolic and bariatric surgery all mortalities by the MBS Committee within the B. Bariatric coordinator required 60-day time limit from the time of discovery. C. All surgeons performing metabolic and bariatric sur8. Qualified metabolic and bariatric surgeon call covergery at the center age: Centers received deficiencies because they failed to D. Operating room nursing representative provide documentation of a bariatric surgery-specific 2. The most common MBSAQIP site survey deficiency is call schedule. Some centers did not provide protocols associated with: outlining the care of an unaffiliated metabolic and barA. Credentialing iatric surgery patient presenting at the center. B. Data entry 9. Metabolic and bariatric surgeon verification: In almost C. Joint Commission accreditation all cases, deficiencies were issued because there was D. Furniture insufficient documentation of the required bariatric-­ 3. The MBSAQIP standard found to have the highest numspecific CME credits. ber of deficiencies is: 10. Facilities, equipment, and instruments: The three most A. Standard 2 – Commitment to Quality Care common deficiencies listed with this standard include B. Standard 3 – Facilities, Equipment, and Instruments absence of appropriate furniture in the waiting areas, C. Standard 6 – Data Collection lack of toilet supports, and the inability of centers to D. Standard 7 – Continuous Quality Improvement document equipment weight limits. 4. What is the estimated number of metabolic and bariatric surgery centers participating in a nationally recognized accreditation program? Conclusion A. 63% B. 74% As the worldwide epidemic of obesity continues its exponenC. 85% tial growth, the demand on surgeons to treat patients safely D. 92% 116 References 1. Hales CM, Fryar CD, Carroll MD, Freedman DS, Ogden CL. Trends in obesity and severe obesity prevalence in US youth and adults by sex and age, 2007-2008 to 2015-2016. JAMA. United States. 2018:1723–5. 2. NIH conference. Gastrointestinal surgery for severe obesity. Consensus Development Conference Panel. Ann Intern Med 1991;115:956–61. 3. Resource Category: Estimate of Bariatric Surgery Numbers|American Society for Metabolic and Bariatric Surgery. 2019. at asmbs.org/ resource-categories/estimate-of-bariatric-surgery-numbers. 4. Birkmeyer JD, Edward E. Mason lecture: strategies for improving the quality of bariatric surgery. Surg Obes Relat Dis. 2013;9: 604–8. 5. English WJ, DeMaria EJ, Brethauer SA, Mattar SG, Rosenthal RJ, Morton JM. American Society for Metabolic and Bariatric Surgery estimation of metabolic and bariatric procedures performed in the United States in 2016. Surg Obes Relat Dis. 2018;14:259–63. 6. Silber JH, Williams SV, Krakauer H, Schwartz JS. Hospital and patient characteristics associated with death after surgery. A study of adverse occurrence and failure to rescue. Med Care. 1992;30:615–29. 7. Alkhenizan A, Shaw C. Impact of accreditation on the quality of healthcare services: a systematic review of the literature. Ann Saudi Med. 2011;31:407–16. 8. Desveaux L, Mitchell JI, Shaw J, Ivers NM. Understanding the impact of accreditation on quality in healthcare: a grounded theory approach. Int J Qual Health Care. 2018;30:241. 9. Inabnet WB 3rd, Bour E, Carlin AM, et al. Joint task force recommendations for credentialing of bariatric surgeons. Surg Obes Relat Dis. 2013;9:595–7. 10. American Society for Bariatric Surgery’s guidelines for granting privileges in bariatric surgery. Surg Obes Relat Dis. 2006;2:65–7. 11. Clements R, Saber A, Teixeira J, Provost D, Fanelli R, Richardson W. Guidelines for institutions granting bariatric privileges utilizing laparoscopic techniques. Society of American Gastrointestinal and Endoscopic Surgeons guidelines committee. Surg Endosc. 2011;25:671–6. 12. Schirmer B, Jones DB. The American College of Surgeons bariatric surgery center network: establishing standards. Bull Am Coll Surg. 2007;92:21–7. 13. Garg T, Birge K, Ulysses R, Azagury D, Rivas H, Morton JM. A postoperative nutritional consult improves bariatric surgery outcomes. Surg Obes Relat Dis. 2016;12:1052–6. 14. Kushner RF, Neff LM. Bariatric surgery: a key role for registered dietitians. J Am Diet Assoc. 2010;110:524–6. 15. Beckman L, Earthman C. Nutritional implications of bariatric surgery and the role of registered dietitians. J Acad Nutr Diet. 2013;113:398–9. 16. Henrickson HC, Ashton KR, Windover AK, Heinberg LJ. Psychological considerations for bariatric surgery among older adults. Obes Surg. 2009;19:211–6. 17. Kushner RF, Sarwer DB. Medical and behavioral evaluation of patients with obesity. Psychiatr Clin North Am. 2011;34: 797–812. 18. Litz M, Rigby A, Rogers AM, Leslie DL, Hollenbeak CS. The impact of mental health disorders on 30-day readmission after bariatric surgery. Surg Obes Relat Dis. 2018;14:325–31. 19. Blackburn AN, Hajnal A, Leggio L. The gut in the brain: the effects of bariatric surgery on alcohol consumption. Addict Biol. 2017;22:1540–53. W. J. English et al. 20. Mitchell JE, Crosby R, de Zwaan M, et al. Possible risk factors for increased suicide following bariatric surgery. Obesity (Silver Spring). 2013;21:665–72. 21. Castaneda D, Popov VB, Wander P, Thompson CC. Risk of suicide and self-harm is increased after bariatric surgery-a systematic review and meta-analysis. Obes Surg. 2019;29:322–33. 22. Backman O, Stockeld D, Rasmussen F, Naslund E, Marsk R. Alcohol and substance abuse, depression and suicide attempts after roux-en­Y gastric bypass surgery. Br J Surg. 2016;103:1336–42. 23. American medical association house of delegates recognition of obesity as a disease – resolution: 420. 2013. at https://media.npr. org/documents/2013/jun/ama-resolution-obesity.pdf. 24. Merrill E, Grassley J. Women’s stories of their experiences as overweight patients. J Adv Nurs. 2008;64:139–46. 25. Schumann R, Jones SB, Cooper B, et al. Update on best practice recommendations for anesthetic perioperative care and pain management in weight loss surgery, 2004-2007. Obesity (Silver Spring). 2009;17:889–94. 26. Giusti V, De Lucia A, Di Vetta V, et al. Impact of preoperative teaching on surgical option of patients qualifying for bariatric surgery. Obes Surg. 2004;14:1241–6. 27. Beck NN, Johannsen M, Stoving RK, Mehlsen M, Zachariae R. Do postoperative psychotherapeutic interventions and support groups influence weight loss following bariatric surgery? A systematic review and meta-analysis of randomized and nonrandomized trials. Obes Surg. 2012;22:1790–7. 28. Finks JF, Kole KL, Yenumula PR, et al. Predicting risk for serious complications with bariatric surgery: results from the Michigan bariatric surgery collaborative. Ann Surg. 2011;254:633–40. 29. Ramanan B, Gupta PK, Gupta H, Fang X, Forse RA. Development and validation of a bariatric surgery mortality risk calculator. J Am Coll Surg. 2012;214:892–900. 30. Gupta PK, Franck C, Miller WJ, Gupta H, Forse RA. Development and validation of a bariatric surgery morbidity risk calculator using the prospective, multicenter NSQIP dataset. J Am Coll Surg. 2011;212:301–9. 31. Benotti P, Wood GC, Winegar DA, et al. Risk factors associated with mortality after roux-en-Y gastric bypass surgery. Ann Surg. 2014;259:123–30. 32. Thompson LA, Dawson K, Ferdig R, et al. The intersection of online social networking with medical professionalism. J Gen Intern Med. 2008;23:954–7. 33. Farnan JM, Snyder Sulmasy L, Worster BK, Chaudhry HJ, Rhyne JA, Arora VM. Online medical professionalism: patient and public relationships: policy statement from the American College of Physicians and the Federation of State Medical Boards. Ann Intern Med. 2013;158:620–7. 34. Pratt JSA, Browne A, Browne NT, et al. ASMBS pediatric metabolic and bariatric surgery guidelines, 2018. Surg Obes Relat Dis. 2018;14:882–901. 35. Inge TH, Zeller MH, Jenkins TM, et al. Perioperative outcomes of adolescents undergoing bariatric surgery: the teen-longitudinal assessment of bariatric surgery (teen-LABS) study. JAMA Pediatr. 2014;168:47–53. 36. Zeitler P, Hirst K, Pyle L, et al. A clinical trial to maintain glycemic control in youth with type 2 diabetes. N Engl J Med. 2012;366:2247–56. 37. Ibele AR, Mattar SG. Adolescent bariatric surgery. Surg Clin North Am. 2011;91:1339–51. 38. Provost D, Jones D, English W, et al. Lessons learned from metabolic and bariatric surgery accreditation and quality improvement program (MBSAQIP) site surveys: Most common deficiencies and corrective actions – science direct. Surg Obes Relat Dis. 2017:S13. Evaluation of Preoperative Weight Loss 10 Hussna Wakily and Aurora D. Pryor programs to decrease body mass index (BMI) would theoretically result in decreased perioperative and postoperative complications such as bleeding, wound infection, etc. In 1. To explain the principles of preoperative weight addition, shorter operative times and hospital stays could loss occur. In addition to the hypothetical risk improvement with 2. To review the data supporting and refuting the benpreoperative weight loss, many medical providers theorized efits of preoperative weight loss that patients who are able to demonstrate PWL are more motivated and serious about adhering to postoperative diet and exercise recommendations. Based on these theoretical advantages, many physicians have supported PWL [4]. In Introduction addition, over the last several years, many insurance compaObesity has become an epidemic in the USA and around the nies added attempted PWL as a prerequisite for surgery [1]. world, leading to increased interest in bariatric surgery as a Many surgeons have questioned, however, if it is truly approtreatment option. Many bariatric patients have had multiple priate to exclude patients from definitive therapy if they failed attempts at weight loss and are looking for longer-­ prove that medical management is ineffective [5]. Most PWL programs include several meetings with nutrilasting results. Although there is consensus for many surgical procedures, optimal preoperative management is more tionists and physicians, as well as psychiatric assessments debated. Preoperative weight loss (PWL) has been proposed and weigh-ins at these appointments. If the patients are as a screening tool for predicting success in surgical candi- unable to show adequate weight loss or if they miss appointdates. It has also been mandated by many surgeons and ments, they are forced to start over or are even refused surinsurers [1]. This chapter will discuss the strategy of PWL gery. The guidelines used by some surgeons and insurance carriers originated with the National Institutes of Health and review the available evidence. (NIH) Consensus Development Conference on Gastrointestinal Surgery for Severe Obesity [6] convened in 1995 and published in 1998 [7]. This consensus group based Principle Behind the Support of Preoperative the recommendation for attempted weight loss on a review of Weight Loss Medline queryable published reports. They supported It is an accepted concept that patients who weigh less have attempted medical weight loss as some patients may be sucdecreased risk with surgery and less weight-related comor- cessful with diet and exercise alone; however, no data was bidity. Thinner patients generally require less rigorous pre- presented to demonstrate differences with outcomes after operative clearance than their heavier counterparts due to a weight loss surgery with or without pre-op diet attempts. In lower burden of obesity-related disease [2]. In addition, it is fact, the NIH panel concluded that less than 20% of patients technically easier to operate on someone who is thinner due have long-term success with diet and exercise alone, bringto improved exposure and accessibility [3]. Preoperative ing into question the legitimacy of the proposed 6-month PWL requirement. Since the NIH consensus panel, many authors have studH. Wakily ied the impact of mandated PWL on eventual surgery. Jamal Division of General Surgery, Department of Surgery, NY and NJ Surgical Associates, Queens, NY, USA and colleagues compared two groups of patients: 72 undergoing a mandated 13-week dietary counseling program and A. D. Pryor (*) Department of Surgery, Stony Brook Medicine, 252 without this requirement. Both groups were similar preStony Brook, NY, USA operatively except for a slight difference in sleep apnea. The Chapter Objectives e-mail: aurora.pryor@stonybrookmedicine.edu © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_10 117 118 PWL group had a 50% higher dropout rate before surgery (28% versus 19%), lower percentage of excess weight loss (%EWL), and higher BMI and weight following surgery. Other tracked outcomes were equivalent [8]. Sadhasivam and colleagues performed a retrospective review of their bariatric surgery candidates from 2001 to 2005 and looked at reasons patients did not undergo surgery. They reviewed 1054 patients and found that only 519 (49%) underwent bariatric surgery and that 29.7% were denied due to insurance denial. Major denial reasons were that patients could not meet the strict preoperative prerequisites established, such as 5–10% reduction in BMI, documented prior weight loss attempts, multiple meetings with nutritionist, etc. Other reasons patients did not undergo surgery were because of frustration with the rigorous requirements and long waiting time. From 2001 to 2005, the authors demonstrated an increase in insurance denials from 9.9% to 19.9% [9]. This study is important in showing that insurance company mandates are resulting in increasing denials for bariatric surgery coverage. eview of the Data Supporting and Refuting R the Benefit of Preoperative Weight Loss Postoperative Weight Loss The Obesity Surgery Mortality Risk Score has been used as a strategy to predict perioperative mortality risk [2]. It is a multivariable analysis used to identify BMI >50, hypertension (HTN), male gender, elevated pulmonary embolism risk, and age >45 years as independent risk variables for bariatric surgery [2]. Among those variables, BMI is the only one that can be modified preoperatively, potentially supporting mandated PWL for higher-risk patients. However, several studies addressing PWL as a means for risk reduction have failed to demonstrate a benefit. Harnisch and colleagues compared two groups of patients undergoing Roux-en-Y gastric bypass (RYGB), those achieving greater than 10 lbs. weight loss (88 patients) before surgery and those that instead gained at least 10 lb. (115 patients). The authors failed to find a difference in perioperative complications, comorbidity resolution, or weight loss at 12 months compared to the immediate preoperative weight [10]. Alami and colleagues completed a rare randomized controlled trial study comparing 26 patients completing the required PWL versus 35 with no preoperative diet requirements [11]. Although excess weight loss was noted to be greater in the PWL group compared to the non-PWL at 6 weeks and 3 months, there was no change in 6-month follow-up visit and 1-year H. Wakily and A. D. Pryor postoperative weight loss. In addition, the groups showed no difference in intraoperative complications or conversions, complication rates, blood loss, or hospital stay. In the 12-month follow-up reported by Solomon, there was also no difference in BMI or comorbidity resolution at 1 year in the population as a whole. A small benefit in %EWL at 1 year was seen, however, in the subgroup able to lose more than 5%EWL preoperatively [12]. In 2011, Cassie, Menezes, and Birch reviewed 17 studies, including 4611 patients that showed PWL beneficial, and 20 studies with 2075 patients showing no benefits to PWL [13]. This analysis includes the studies previously discussed in this chapter (Table 10.1). Most reviewed analyses focused on the 12-month follow-up point. Combining all studies reporting 12-month data, the non-preoperative weight loss groups had an estimated weight loss of 70.7 ± 5.7% versus the preoperative weight loss group of 69.0 ± 7.1%. At 2 years, there was still no significant difference in both weight groups with the PWL showing 66.7 ± 2.7% estimated weight loss and the non-preoperative weight loss group at 72 ± 6.3%. The authors concluded that there was inadequate data to support mandated PWL based on the outcome of postoperative weight loss [1, 2]. Livhits and colleagues performed a meta-analysis of 15 articles including 3404 patients. Only two of the included cohorts were excluded from the Cassie review. Not surprisingly, the authors drew similar conclusions. Of the 15 articles analyzed, 5 studies had positive effects for PWL, 2 studies showed positive short-term effects, 5 studies showed no difference, and 1 study showed a negative effect. Overall no significant heterogeneity was seen among the studies with results of postoperative weight loss [3]. Operative Time One of the hypothesized benefits of PWL includes shorter operative times, so looking at the studies that analyzed, this is important in determining if PWL is needed. In the Alami study, total operating time was greater in the non-­preoperative weight loss group (257.6 ± 27.8 min versus 220.2 ± 31.5 min) as compared with the preoperative weight loss group [4]. Harnisch and colleagues also found a slight benefit in OR time with PWL (119.7 versus 104.9 min, P = 0.02) [5]. The savings of 12.5–23 min with PWL are consistent in other studies as well [1, 3]. The problem with these papers is that it is not properly distinguished how operating time is measured, so there could be a discrepancy in what is a standardized operating time. Also, there is no study so far that has demonstrated that the time saved has contributed to improved patient safety and outcomes. 10 Evaluation of Preoperative Weight Loss 119 Table 10.1 Studies comparing effectiveness of preoperative weight loss Study inclusion Investigator Year 2007 Alami 2008 2007 2005 2010 2009 2007 2008 2008 Study type RCT Patients (n) 61 Procedure LRYGB Alger-Mayer Ali Alvarado Becouarn Benotti Broderick-­Villa Carlin Conlee Prospective Retrospective Retrospective Retrospective Retrospective Retrospective Retrospective Retrospective 150 351 90 507 881 353 295 105 RYGB LRYGB LRYGB RYGB/LAGB/SG LRYGB/RYGB RYGB LRYGB RYGB 2010 1997 2008 2008 2008 Eisenberg Finigan Fujioka Gallo Harnisch Retrospective Prospective Retrospective Retrospective Retrospective 256 31 121 494 203 LRYGB LAGB LRYGB/RYGB LAGB LRYGB 2005 2008 Hong Huerta Retrospective Retrospective 100 40 LAGB RYGB 2009 2005 1995 2007 2008 Jantz Liu Martin Micucci Mrad Retrospective Retrospective Prospective Retrospective Retrospective 384 95 100 NR 146 2005 2008 Phan Reiss Retrospective Retrospective 364 262 LRYGB LRYGB RYGB RYGB LRYGB/RYGB/ LAGB/VGB LRYGB/RYGB/LAGB LRYGB 2009 2009 2007 1999 Segaran Solomon Still Van de Weijgert Prospective RCT Prospective Retrospective 37 44 884 153 RYGB/LAGB/SG LRYGB LRYGB/RYGB RYGB/VGB Variables assessed Postoperative EWL, operating room time, complication rate, comorbidity resolution Postoperative EWL Postoperative EWL Postoperative EWL, operating time, comorbidity resolution Postoperative EWL Complication rate Postoperative EWL Postoperative EWL Postoperative EWL, operating time, complication rate, length of stay Postoperative EWL Postoperative EWL Postoperative EWL, complication rate Postoperative EWL, operating time Postoperative EWL, operating time, complication rate, comorbidity resolution Postoperative EWL Postoperative EWL, operating time, complication rate, length of stay Postoperative EWL Operating time, complication rate, length of stay Postoperative EWL, complication rate Postoperative EWL Postoperative EWL Postoperative EWL Postoperative EWL, operating time, complication rate, length of stay Complication rate Postoperative EWL, conversion rate, complication rate Postoperative weight loss, length of stay Postoperative EWL Adapted from Cassie et al. [1] RCT randomized controlled trial, LRYGB laparoscopic Roux-en-Y gastric bypass, EWL excess weight loss, RYGB Roux-en-Y gastric bypass, LAGB laparoscopic adjustable gastric banding, SG sleeve gastrectomy, VGB vertical gastric banding Operative Complications Livhits and colleagues proposed that meta-analysis of complications is difficult due to the lack of consistency in definition. However, they concluded that there was no significant difference between groups [3]. Cassie noted decreased complications with PWL in 2 of 11 studies. However, when the data from the reviewed papers were pooled, the complication rates for the preoperative weight loss group were 18.8 ± 10.6% versus 21.4 ± 13.1% in the non-preoperative weight loss group showing that there was no real difference in the two groups [1]. Hospital Stay Decreased length of stay is one benefit that would also result in cost-reducing measures, but again there are inadequate data to support a length of stay benefit with PWL. Cassie reported on five studies with the length of stay ranging from 2.2 to 4.3 days for the PWL versus 2.3–6.0 days for the non-­ preoperative weight loss group. The mean from pooled studies was 3.34 ± 0.83 for the PWL versus 3.98 ± 1.49 days for the non-preoperative weight loss group [1]. Although there was a trend in support of PWL, the data are inconclusive. Liver Reduction and VLED Many bariatric surgeons have recognized variability in fatty liver intraoperatively and attribute reduced liver size to preoperative weight loss. Some surgeons have specifically implemented a very-low-energy diet (VLED) for weight loss and liver volume reduction. Colles investigated the actual changes in liver volume and the pattern of this change with 120 H. Wakily and A. D. Pryor Baseline Week 12 Fig. 10.1 Single cross-sectional images of the liver performed by computed tomography at baseline and week 12 of a very-low-energy diet. The images, taken from within a series of contiguous 8-mm slices used to calculate total liver volume, illustrate the extent of the change in liver volume with weight loss in a 35-year-old man with an initial liver volume of 3.7 L and final liver volume of 2.4 L. A 35% reduction in liver size and a weight loss of 18 kg were observed VLED, the relative change in liver volume, body weight and visceral and subcutaneous adipose tissue (VAT/SAT) areas, the clinical and biochemical risk factors that may predict an enlarged pretreatment liver or predict the total change in liver volume after treatment, patient acceptability and compliance, and treatment side effects. Patients with BMI between 40 and 50 were chosen based on the fact that surgeons find that patients in this range posed the greatest surgical difficultly. The diet itself consisted of three shakes/day providing 456 kcal, 52 g protein, 7 g fat, and 45 g carbohydrate plus the recommended daily allowance of vitamins, minerals, and trace elements. This diet took place over 12 weeks. The changes in the liver were assessed by computed tomography (CT) scan and magnetic resonance imaging (MRI) during the 12-week interval (Fig. 10.1). At the end of the diet, the average decrease in liver size was 18.7%. Also, this study showed that the larger the initial liver volume, the better the decrease in liver volume. The authors reported no hepatomegaly in the study patients and a low perioperative risk, although the study was not powered for complications. The most important pattern noted is that the majority of the volume reduction occurred within the first 2 weeks [6]. This data supports potential benefit with PWL; however, a short 2-week time course may be adequate. Healthcare System became a great source for evaluating this specific type of population. Collins and colleagues looked at changes in liver volume and adipose tissue in patients with BMI >50 [7]. The authors felt these patients in theory would benefit most from a reduction in liver volume and adipose tissue to facilitate surgery and decrease complication rates. The comorbidities with increased prevalence in the super obese include all those associated with obesity: type II diabetes mellitus, hypertension, obstructive sleep apnea, congestive heart failure, and so on. The technical difficulties related to this population of patients include thick abdominal wall, excess visceral adipose tissue, a relatively short mesentery, and an enlarged fatty liver. Altering these characteristics should in theory decrease complication rates and shorten operative times. To achieve acute preoperative weight loss, Collins employed a liquid low-calorie diet consisting of 800 kcal/ day, nutritional and behavioral counseling, and education with the goal of a 10% weight loss reduction. The goal of this study was to evaluate change in obesity-related comorbidities, liver volume, and subcutaneous adipose tissue over several weeks. Since the weight loss and methods were drastic, the patients were required to have multiple assessments during their preoperative course. The authors also tracked changes in mediations, weekly laboratory tests, and weekly physical exams because of the aggressive nature of the weight loss. At baseline and after the completion of the diet program, computed tomography (CT) scans of the abdomen were compared. A total of 30 patients underwent the program with a mean BMI at baseline of 56, mean age 53. The program lasted a total of 9 weeks, and the patients had an Super Obese Characteristics that put patients at a higher level of risk for bariatric surgery include BMI >50, male gender, and a lower socioeconomic status. The Veterans Affairs Pittsburgh 10 Evaluation of Preoperative Weight Loss average body weight loss of 12.1%. After the low-calorie diet was completed, the mean BMI decreased to 49. They recognized an 18% decrease in liver volume over 9 weeks in patients enrolled in the study. They did not, however, ­compare complications or outcomes to patients not completing the diet. Their final conclusion is that a preoperative diet is safe to decrease liver volume for bariatric surgery in an effort to improve perioperative and postoperative course, especially for those with a BMI >50. As with Colles and Collins, other studies have supported this finding as well [8]. Additional Considerations with PWL 121 scored greater in the dietary adherence experienced a weight loss that was 2.4% points greater at postoperative week 40 and 3.8% points greater at week 66 compared to those patients with a lower score on the dietary adherence. By week 92, both groups had regained some of their weight, but the group that scored in the high adherence group still had achieved a weight loss that was 4.5% points greater than those in the low adherence group, representing a 28% greater weight loss. This study was able to demonstrate improvements in psychosocial status postoperatively. They had improved self-esteem, increased positive affect, and decreased negative affect and depressive symptoms, as well as had changes in their eating habits [13]. These results suggest that there is a positive psychosocial benefit to nutritional and behavioral education, although not necessarily PWL. Teaching better eating habits not only helps the patients maintain postoperative weight loss but also improves their self-esteem. All these translate to patients who participate in a preoperative educational program and may be better equipped to help keep off weight postoperatively. As a result, Sarwer supports that PWL should be encouraged but not used as a way to screen for surgical candidates who would obtain the most from bariatric surgery. Most bariatric patients gain some portion of their weight back after their postoperative nadir. It is supported by most surgeons that weight regain and weight loss failure are largely tied to behavioral and psychosocial causes, although recent research suggests that the root cause of weight regain may be due to the genetic predisposition [9] or from alteration of the physiological mechanisms by which metabolic operations appear to work [10–12]. The psychosocial benefit of PWL may have the most dramatic results on maintaining postoperative weight loss in the long term. Sarwer analyzed the relationship between preoperative eating behavior and postoperative dietary adherence Study Limitations [13]. The goal of this study was to look at the relationship of postoperative weight loss to preoperative psychosocial vari- The studies reviewed in this chapter have some limitations to ables such as self-esteem and mood, as well as preoperative address. Only one study is available with a prospective, raneating behaviors, dietary intake, and patients’ self-reported domized study design [4]; however, even in this analysis, adherence to the postoperative diet over a 92-week period. fewer than 100 patients were available. The other studies are Approximately 200 patients participated in the study, with primarily retrospective studies with significant variability in the initial evaluation at 4 weeks before surgery. The partici- data collection. Larger-scale, prospective randomized trials pants completed a psychosocial/behavioral evaluation to would need to support a benefit in PWL before this should be assess their appropriateness for surgery followed by a packet mandated. The Sarwer study was limited due to the high of questionnaires. They were again asked to fill out the ques- number of attrition, which is typical for these types of studtionnaires at 20, 40, 66, and 92 weeks after surgery. The ies, and only a 2-week educational period prior to surgery. packet included the following measures: Rosenberg Self-­ Only 56% of the patients completed the study, impacting the Esteem Scale, Beck Depression Inventory-II, Positive and significance of the paper by decreasing the population size. Negative Affect Scale, Eating Inventory, Block 98 Food Another limitation to this paper was that the data was based Frequency Questionnaire, dietary adherence, and weight. on self-analysis, which is difficult to interpret [13]. Approximately 2 weeks prior to surgery, the participants met with a dietician and were instructed on dietary and behavioral changes that the authors felt would give them the best ASMBS Position Statement postoperative outcome, followed by dietary instructions postoperatively to help maintain the weight loss. From the The American Society for Metabolic and Bariatric Surgery variables that were looked at, the ones that were significant (ASMBS) issued a position statement due to questions raised predictors for success (measured by percentage of weight to the society by physicians, hospitals, patients, and insurloss over time) included gender, baseline cognitive restraint, ance companies regarding preoperative weight loss [14]. and self-reported dietary adherence. Those patients who (This statement is reviewed in Chap. 11.) The final summary were able to show cognitive restraint preoperatively also did and recommendations, as also demonstrated by the studies well at 20 weeks postoperatively, when the patients were reviewed here, are that there is a low level of evidence supadvanced back to a regular diet. Those individuals who porting the need for preoperative weight loss. Class I or 122 evidence-­based studies currently are not available to support the request for mandated PWL. Lower levels of evidence do exist, but the data and results are not consistent. Although there is some evidence in the Class II–IV range for acute preoperative weight loss, the studies are not consistent, and some are conflicting, leaving us with no clear answers. There is some low level of evidence to suggest that preoperative weight loss can help with evaluating a patient’s adherence to the new lifestyle, but this should be judged by each individual situation. The studies so far have shown no difference in comorbidity improvement, postoperative complications, or postoperative weight loss to justify the need for PWL. The current recommendation is that insurance companies and physicians should reevaluate the need for PWL. Thus far, it contributes to higher attrition rates, increases frustration among patients, and blocks certain patients from obtaining a life-altering and beneficial surgery. Conclusion The only evidence-based support for preoperative education and encouraging PWL is that these tools may help patients prepare for surgery and understand techniques to maintain a healthier lifestyle postoperatively. The data so far is inconsistent and inconclusive with regard to the benefits of PWL, but the majority of the studies show no difference in complication rates or morbidities in patients undergoing PWL versus no PWL. If considering PWL, the data supports the notion that shorter-term preoperative diets (i.e., 2 weeks) may also reduce liver volume and reduce operative time, without significant reduction in risk. Mandated programs in excess of this are a barrier to our patients and without supportive evidence. Question Section 1. Studies have shown that a very-low-energy diet has been beneficial and should be started preoperatively. What is the optimal timing for starting the diet? A. 2 weeks B. 6 weeks C. 8 weeks D. 12 weeks 2. The ASMBS position statement was issued to clarify the validity of PWL by reviewing the current literature and evidence. The statement’s final conclusion is that there is a strong level of evidence to justify the use of PWL. A. True B. False H. Wakily and A. D. Pryor References 1. Cassie S, Menezes C, Birch DW, Shi X, Karmali S. Effect of preoperative weight loss in bariatric surgical patients: a systematic review. Surg Obes Relat Dis. 2011;7(6):760–7. 2. Ray EC, Nickels MW, Sayeed S, Sax HC. Predicting success after gastric bypass: the role of psychosocial and behavioral factors. Surgery. 2003;134(4):555–63.. discussion 563–4 3. Livhits M, Mercado C, Yermilov I, Parikh JA, Dutson E, Mehran A, et al. Does weight loss immediately before bariatric surgery improve surgical outcomes: a systematic review. Surg Obes Relat Dis. 2009;5(6):713–21. 4. Alami RS, Morton JM, Schuster R, Lie J, Sanchez BR, Peters A, et al. Is there a benefit to preoperative weight loss in gastric bypass patients? A prospective randomized trial. Surg Obes Relat Dis. 2007;3(2):141–5.. discussion 145–6 5. Harnisch MC, Portenier DD, Pryor AD, Prince-Petersen R, Grant JP, DeMaria EJ. Preoperative weight gain does not predict failure of weight loss or co-morbidity resolution of laparoscopic roux-­ en-­ y gastric bypass for morbid obesity. Surg Obes Relat Dis. 2008;4(3):445–50. 6. Colles SL, Dixon JB, Marks P, Strauss BJ, O’Brien PE. Preoperative weight loss with a very-low-energy diet: quantitation of changes in liver and abdominal fat by serial imaging. Am J Clin Nutr. 2006;84(2):304–11. 7. Collins J, McCloskey C, Titchner R, Goodpaster B, Hoffman M, Hauser D, et al. Preoperative weight loss in high-risk superobese bariatric patients: a computed tomography-based analysis. Surg Obes Relat Dis. 2011;7(4):480–5. 8. Fris RJ. Preoperative low energy diet diminishes liver size. Obes Surg. 2004;14(9):1165–70. 9. Matzko ME, Argyropoulos G, Wood GC, Chu X, McCarter RJ, Still CD, et al. Association of ghrelin receptor promoter polymorphisms with weight loss following Roux-en-Y gastric bypass surgery. Obes Surg. 2012;22(5):783–90. 10. Liou AP, Paziuk M, Luevano JM Jr, Machineni S, Turnbaugh PJ, Kaplan LM. Conserved shifts in the gut microbiota due to gastric bypass reduce host weight and adiposity. Sci Transl Med. 2013;5(178):178ra41. 11. Angelakis E, Armougom F, Million M, Raoult D. The relationship between gut microbiota and weight gain in humans. Future Microbiol. 2012;7(1):91–109. 12. Frazier TH, DiBaise JK, McClain CJ. Gut microbiota, intestinal permeability, obesity-induced inflammation, and liver injury. JPEN J Parenter Enteral Nutr. 2011;35(5 Suppl):14S–20. 13. Sarwer DB, Wadden TA, Moore RH, Baker AW, Gibbons LM, Raper SE, et al. Preoperative eating behavior, postoperative dietary adherence and weight loss after gastric bypass surgery. Surg Obes Relat Dis. 2008;4(5):640–6. 14. Brethauer S. ASMBS position statement on preoperative supervised weight loss requirements. Surg Obes Relat Dis. 2011;7(3):257–60. ASMBS Position Statements 11 Stacy A. Brethauer and Xiaoxi (Chelsea) Feng Chapter Objectives 1. Review bariatric surgery recommendations based on current knowledge, expert opinion, and published peer-reviewed scientific evidence. 2. Summarize current position statements of the ASMBS for the care and treatment of bariatric patients. Introduction Why Have Position Statements? Most large medical and surgical societies publish position statements and guidelines for their membership. The majority of these position statements are evidence-based documents that provide a comprehensive summary of the available data on a topic that is of particular interest to the membership. The intent of publishing these statements in many cases is to clarify a controversial issue, to provide guidance for healthcare leaders and payers, and to provide support for clinical decisions made by the membership. The American Society for Metabolic and Bariatric Surgery (ASMBS) publishes position statements in response to numerous inquiries made to the society by patients, physicians, society members, hospitals, and others regarding a particular topic relevant to the field. In these statements, available data are summarized, and recommendations for treatment are made based on current knowledge, expert opinion, and published peer-reviewed scientific evidence available at the time. The ASMBS leadership also collaborates S. A. Brethauer (*) Department of Surgery, The Ohio State University, Columbus, OH, USA e-mail: Stacy.Brethauer@osumc.edu with other medical societies to develop and publish practice guidelines that are relevant to the care of the bariatric surgery patient [1]. How Are Statements Developed? ASMBS statements can be generated within the Clinical Issues Committee (CIC) or within other committees as the need arises. The Executive Council can also task the CIC to generate position statements based on the need to clarify the society’s stance on a particular topic. The Clinical Issues Committee has formalized the processes for statement development and approval to ensure that these documents are developed, published, and revised in the appropriate time frame. The development and vetting process for position statements are summarized in Fig. 11.1. Summary of Current Position Statements Access to Care for Obesity Treatment 1. Obesity is a medical condition that meets all criteria as a disease, including a genetic predisposition and personal, societal, and environmental factors that contribute to expression of the disease [2]. 2. Obesity is an independent risk factor for heart disease and is predominant in patients with hypertension, high cholesterol, and type 2 diabetes. 3. People who suffer from the disease of obesity should be free from prejudice and discrimination in accessing care for obesity. 4. People who suffer from the chronic disease of obesity should have access to evidence-based medical, pharmaceutical, behavioral, and surgical care similar to the access provided for the evaluation and management of other chronic disease. X. (C.) Feng Department of General Surgery, Cleveland Clinic, Cleveland, OH, USA © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_11 123 124 S. A. Brethauer and X. (C.) Feng 5. Prevention of obesity through new strategies of healthy lifestyles should complement the commitment to treat those currently suffering from the metabolic consequences of the disease. 6. Weight loss surgery is effective, durable, and increases quality and quantity of life. Obesity should be treated. reoperative Supervised Weight Loss P Requirements 1. There are no data from any randomized controlled trial, large prospective study, or meta-analysis to support the practice of insurance-mandated preoperative weight loss. The discriminatory, arbitrary, and scientifically unfounded practice of insurance-mandated preoperative weight loss contributes to patient attrition, causes unnecessary delay of lifesaving treatment, leads to the progression of life-­ threatening comorbid conditions, is unethical, and should be abandoned [3]. 2. There is no level 1 data in the surgical literature or consensus in the medical literature (based on over 40 published RCTs) that has clearly identified any 1 dietary regimen, duration, or type of weight loss program that is optimal for patients with clinically severe obesity [3]. 3. Patients seeking surgical treatment for clinically severe obesity should be evaluated based on their initial BMI and comorbid conditions. The provider is best able to determine what constitutes failed weight loss efforts for their patient [3]. Fig. 11.1 (a, b) The general algorithm for statement development within the ASMBS a I mpact of Obesity and Obesity Treatment on Fertility and Fertility Therapy There is a very high prevalence of obesity among women of childbearing age. Obesity in women is associated with an increased risk of infertility and an increased rate of complications during every stage of pregnancy. Obesity is associated with PCOS and IR, which also negatively impact fertility. Overall, however, there is a paucity of high-level evidence regarding the impact of obesity and obesity treatment on fertility and infertility treatment. Ongoing investigation and randomized controlled trials are necessary to fully understand the role of obesity and the impact of medical and surgical treatments for obesity on male and female fertility and infertility treatment outcomes [4]. 1. Obesity is associated with a significant delay in conception that is partly, but not entirely, due to an impact on normal ovulation. 2. Obesity reduces male fertility parameters and should be considered in the evaluation of a couple presenting with infertility. 3. The symptoms of PCOS, particularly with respect to fertility and metabolic disturbance, are exacerbated in the presence of obesity. 4. Weight loss can improve weight-associated causes for infertility such as PCOS and IR. 5. For some overweight and obese women, particularly with PCOS, weight loss may improve ovulatory function, leading to improved fertility. 11 ASMBS Position Statements b 125 Alcohol Use Before and After Bariatric Surgery There is conflicting data as to the lifetime and current prevalence of alcohol use disorder (AUD) in patients seeking weight loss surgery. Most studies indicate that AUD affects a minority of bariatric surgery patients. Studies have shown that some individuals are at risk for AUD relapse or for developing new-onset AUD after weight loss surgery, especially after gastric bypass. Other studies have shown a decrease in high-risk drinking after surgery compared with baseline [5]. Based on current studies, gastric bypass surgery is associated with: 1. Accelerated alcohol absorption (shorter time to reach maximum concentration) 2. Higher maximum alcohol concentration 3. Longer time to eliminate alcohol in both men and women 4. Increased risk for development of AUD Fig. 11.1 (continued) 6. Obese women have a lower probability of achieving live birth after in vitro fertilization. 7. Bariatric surgery is effective in achieving significant and sustained weight loss in morbidly obese women and has been shown in case-control studies to improve fertility. 8. Pregnancy is not recommended during the rapid weight loss phase after bariatric surgery; therefore, counseling and follow-up regarding contraception during this period are important. 9. The specific impact of either medical weight loss treatments or bariatric surgery on the responsiveness to subsequent treatments for infertility in both men and women is not clearly understood at this time. This position statement has been endorsed by the American College of Obstetricians and Gynecologists, May 2017, and should be construed as its clinical guidance. The data are less clear regarding altered pharmacokinetics after sleeve gastrectomy, and there is no evidence that alcohol absorption is affected by gastric banding. Given the recent increase in popularity of sleeve gastrectomy, more studies regarding the pharmacokinetic effects of sleeve gastrectomy on alcohol metabolism are needed. Patients undergoing bariatric surgery should be screened and educated regarding alcohol intake both before and after surgery. Active AUD is considered a contraindication by most programs and in published guidelines [1]. Adequate screening, assessment, and preoperative preparation may help decrease the risk of AUD in bariatric surgery patients [6]. A period of sustained abstinence with treatment is indicated before weight loss surgery. A history of AUD is not a contraindication to bariatric surgery. However, patients should be made aware that AUD can occur in the long term after bariatric surgery. ariatric/Metabolic Surgery in Class I Obesity B (BMI 30–35kg/m2) There is a growing body of literature supporting the use of metabolic surgery for patients with lower BMIs, particularly for patient with type 2 diabetes mellitus (T2DM). The current ASMBS position statement provides a summary and recommendations based on the available evidence [7]: 1. Class I obesity (BMI 30–35 kg/m2) causes or exacerbates multiple other diseases, decreases longevity, and impairs quality of life. Patients with class I obesity require durable treatment for their disease. 2. Current nonsurgical treatments for class I obesity are often ineffective at achieving major, long-term weight reduction and resolution of comorbidities. 126 S. A. Brethauer and X. (C.) Feng 3. The existing BMI inclusion criterion of ≥35 kg/m2 as a 3. Patients who have documented moderate to severe OSA prerequisite for bariatric and metabolic surgery—excludshould be strongly encouraged to accept treatment preoping individuals with class I obesity—was established eratively with CPAP and to use it postoperatively until arbitrarily more than a quarter century ago, in the era of clinical evaluation demonstrates resolution of OSA. open surgery when morbidity and mortality of surgery 4. These patients should also bring their CPAP machines, or were significantly higher than today. There is no current at least their masks, with them at the time of surgery and evidence of clinical efficacy, cost-effectiveness, ethics, or use them following bariatric surgery at the discretion of equity that justifies this group being excluded from lifethe surgeon. saving surgical treatment. Access to bariatric and meta- 5. All commonly performed bariatric operations that have bolic surgery should not be denied solely based on this been assessed for impact on OSA have shown evidence outdated threshold. for significant relief of subjective symptoms of OSA and 4. For patients with BMI 30–35 kg/m2 and obesity-related improvement of objective parameters of OSA that may comorbidities who do not achieve substantial, durable not always correlate with amount of weight lost. weight loss, and comorbidity improvement with reason- 6. Since bariatric surgery produces many improvements in able nonsurgical methods, bariatric surgery should be the quality of life and other coexisting medical conditions offered as an option for suitable individuals. In this popufor severely obese patients with OSA, it should be considlation, surgical intervention should be considered after ered as the initial treatment of choice for OSA in this failure of nonsurgical treatments. patient population as opposed to surgical procedures 5. Particularly given the presence of high-quality data in directed at the mandible or tissues of the palate. patients with type 2 diabetes, bariatric and metabolic sur- 7. Routine pulse oximetry or capnography for postoperative gery should be strongly considered for patients with BMI monitoring of patients with OSA after bariatric surgery 30–35 kg/m2 and type 2 diabetes. should be utilized, but the majority of these patients do not routinely require an ICU setting. 6. AGB, SG, and RYGB have been shown to be well-­ tolerated and effective treatments for patients with BMI 8. No clear guidelines exist upon which to base recommendations for retesting for OSA following bariatric surgery. 30–35 kg/m2. Safety and efficacy of these procedures in However, strong consideration should be given to retestlow-BMI patients appear to be similar to results in patients ing patients who present years after bariatric surgery with with severe obesity. regain of weight, a history of previous OSA, and who are 7. Perioperative and long-term nutritional, metabolic, and being reevaluated for appropriate medical and potential nonsurgical support must be provided to patients after preoperative surgical therapy. surgery according to established standards, including the ASMBS Clinical Practice Guidelines. 8. Currently, the best evidence for bariatric and metabolic surgery for patients with class I obesity and comorbid VTE Prophylaxis conditions exists for patients in the 18–65 age group. There is no class I evidence to provide guidance regarding the type, dose, or duration of VTE prophylaxis in the bariatObstructive Sleep Apnea ric surgery patient. Based on current evidence available, the following recommendations are made [9]: Based on the evidence in the literature to date, the following guidelines regarding OSA in the bariatric surgery patient and 1. All bariatric surgery patients are at moderate to high risk its perioperative management are recommended [8]: for VTE events, and VTE prophylaxis should be used. 2. Factors that place patients into a high-risk category for 1. OSA is highly prevalent in the bariatric patient populaVTE after bariatric surgery may include high BMI, tion. The high prevalence demonstrated in some studies advanced age, immobility, prior VTE, known hypercoagsuggests that consideration be given to testing all patients ulable condition, obesity hypoventilation syndrome, puland especially those with any preoperative symptoms monary hypertension, venous stasis disease, hormonal suggesting obstructive sleep apnea. therapy, expected long operative time or open approach, 2. Untreated OSA is yet another comorbidity observed with and male gender. high prevalence in the bariatric patient population that leads 3. Individual practices should develop and adhere to a prototo increased mortality and increased medical disability from col for VTE prevention. Available evidence suggests that several cardiovascular diseases. These observations further adherence to any specific practice for VTE prevention emphasize the value of bariatric surgery as a potentially will reduce but not eliminate VTE as a complication of definitive treatment for OSA in severely obese patients. bariatric surgery. 11 ASMBS Position Statements 127 4. Mechanical prophylaxis is recommended for all bariatric weight loss than is seen after the adjustable gastric band surgery patients. There may be individual circumstances (AGB) or nonsurgical interventions. There is no consis(severe lymphedema) when lower extremity compression tent conclusion on which procedure produces greater devices are not practical and alternative strategies may be weight loss early after surgery. However, most current needed. evidence seems to demonstrate that RYGB produces 5. Early ambulation is recommended for all bariatric surgreater EWL compared to SG beyond the 1st year [13]. gery patients. 2. The effect of SG on GERD is less clear, because GERD 6. The combination of mechanical prophylaxis and chemoimprovement is less predictable and GERD may worsen prophylaxis should be considered based on clinical judgor develop de novo. Preoperative counseling specific to ment and risk of bleeding. Although there is some GERD-related outcomes is recommended for all patients low-level evidence that mechanical prophylaxis alone in undergoing SG. low-risk patient results in low VTE rates (o.4%), the pre- 3. The ASMBS recognizes SG as an acceptable option for a ponderance of bariatric data supports using a combination primary bariatric procedure or as a first-stage procedure of chemoprophylaxis and mechanical prophylaxis with in high-risk patients as part of a planned, staged approach. overall VTE rates o.5%. 4. As with any bariatric procedure, long-term weight regain 7. There are conflicting data in the literature regarding the can occur after SG and may require one or more of a varitype of chemoprophylaxis to use, but the highest-quality ety of re-interventions. data currently available suggest that LMWH offers better 5. Informed consent for SG as a primary procedure should VTE prophylaxis than UFH without increasing the bleedbe consistent with the consent provided for other bariatric ing risk. procedures and, as such, should include the risk of long-­ 8. Most post-discharge VTE events occur within the first term weight regain. In addition, as with all currently rec30 days after surgery. Extended VTE prophylaxis should ognized bariatric procedures, surgeons performing SG are be considered, but there are insufficient data to recomencouraged to prospectively collect, analyze, and report mend a specific dose or duration of extended post-­ their outcome data in peer-reviewed scientific forums. discharge VTE prophylaxis for patients deemed to be at high risk for VTE. 9. The use of IVC filters as the only method of prophylaxis Single-Anastomosis Duodenal Switch before bariatric surgery is not recommended. Filter placement may be considered in combination with chemical The following recommendations are currently endorsed by and mechanical prophylaxis for selected high-risk patient the ASMBS regarding single-anastomosis duodenal switch in whom the risks of VTE are determined to be greater (SADS) for the primary treatment of obesity or metabolic than the risks of filter-related complications. disease [14]: 1. Single-anastomosis duodenal switch procedures are considered investigational at present. The procedure should be performed under a study protocol with third-party There have been three previously published statements by oversight (local or regional ethics committee, institutional the American Society for Metabolic and Bariatric Surgery review board, data monitoring and safety board, clinical(ASMBS) on the use of sleeve gastrectomy (SG) as a bariattrials.gov, or equivalent authority) to ensure continuous ric procedure [10–12]. With the emergence of this procedure evaluation of patient safety and to review adverse events as the most commonly performed, bariatric procedure today and outcomes. prompts a review of the current evidence. Substantial long-­ 2. Publication of short- and long-term safety and efficacy term outcome data published in the peer-reviewed literature, outcomes is strongly encouraged. including studies comparing outcomes of various surgical 3. Data for these procedures from accredited centers should procedures, confirm that SG provides significant and durable be reported to the Metabolic and Bariatric Surgery weight loss, improvements in medical comorbidities, Accreditation and Quality Improvement Program dataimproved quality of life, and low complication and mortality base and separately recorded as single-anastomosis DS rates for obesity treatment [13]. procedures to allow accurate data collection. Sleeve Gastrectomy as a Bariatric Procedure 1. In terms of initial early weight loss and improvement of most weight-related comorbid conditions, SG and RYGB appear similar. Studies demonstrate that SG and Rouen-­Y gastric bypass (RYGB) provide more comparable These recommendations are not intended to impede innovation within our field. The ASMBS understands and supports the need for new and innovative procedures that can further benefit our patient population. The single-­anastomosis 128 modification of the DS procedure, however, represents a significant change to a procedure that is historically rarely performed. In addition to potential unique long-term risks without clear benefit, there are nutritional concerns that will require further evaluation and study to ensure the safety of both our patients and our members and to protect them from undue harm. Gastric Plication Gastric plication is a new bariatric procedure that involves imbricating the anterior or greater curvature of the stomach to reduce gastric volume without placement of a device or gastric resection. This emerging procedure has been performed as a stand-alone procedure as well as a combination procedure with a laparoscopic adjustable gastric band [15]. The ASMBS currently supports the following recommendations regarding gastric plication alone or in combination with adjustable gastric band placement for the treatment of obesity: 1. Gastric plication procedures should be considered investigational at this time. This procedure should be performed under a study protocol with a third-party oversight (local or regional ethics committee, institutional review board [IRB], data monitoring and safety board, or equivalent authority) to ensure continuous evaluation of patient safety and to review adverse events and outcomes. 2. Reporting of short- and long-term safety and efficacy outcomes in the medical literature and scientific meetings is strongly encouraged. Data for these procedures should also be reported to a program’s center of excellence database. 3. Any marketing or advertisement for this procedure should include a statement to the effect that this is an investigational procedure. 4. The ASMBS supports research conducted under an IRB protocol as it pertains to investigational procedures and devices. Investigator meetings held to facilitate research are necessary and supported, as is the reporting of all data through a bariatric quality improvement program or a specific research database. The ASMBS does not support continuing medical education (CME) courses on investigational procedures and devices held for bariatric surgeons for the purpose of use of investigational procedures outside an IRB research protocol. Prevention and Detection of Gastrointestinal Leak There has been a decrease in the incidence of GI leaks after primary stapled open and laparoscopic bariatric procedures S. A. Brethauer and X. (C.) Feng (GB and SG) over time. Despite this decline, a GI leak remains a significant cause of morbidity and mortality and remains a potential complication of these procedures. Early detection and treatment remain pivotal principles in the management of GI leaks and may play a role in reducing subsequent morbidity and mortality. Some of the factors promoting leak may be different between GB and SG procedures. This may be related to the technical differences between the two procedures, as well as the distinct anatomic and physical properties that exist between the sleeve conduit versus the gastric pouch, which are helpful when considering some of the qualitative and temporal dissimilarities reported in the clinical manifestation of GI leaks after these procedures. Despite the advent of new technologies, the management of GI leaks after GB and SG procedures can be extremely complex and involve multiple and/or multimodal treatment options [16]. 1. Intraoperative leak tests (air, dye, endoscopy) are described for both GB and SG. Although widely used, they have not been found to reduce the incidence of leak after GB and SG procedures. 2. Intraoperative leak prevention interventions described for both GB and SG procedures include oversewing, SLR, tissue sealants, and glue. There is still considerable debate over the utility or superiority of any of these interventions. Mandated use of any of these leak prevention interventions was not indicated by the data. 3. Radiographic studies after GB and SG procedures have varying sensitivity and specificity that is affected by study choice, patient factors, facility factors, and reviewer factors. • There is no high-quality evidence available to mandate the routine postoperative use of UGI contrast studies after GB or SG procedures, particularly for SG given the greater likelihood of leaks presenting in a delayed fashion. Routine or selective UGI studies may, however, identify other technical or anatomic problems after GB or SG procedures. Based on current evidence, the decision to perform routine versus selective UGI contrast studies should be left to the discretion of the surgeon, based on factors related to the system of care in place and on other characteristics of the patient and the population being treated. 4. Radiographic evaluation versus surgical exploration for suspected leak after GB and SG. • Clinically unstable patients suspected of having a leak may not be appropriate candidates for radiographic evaluation. Re-exploration through a laparoscopic or open approach should be considered. • In the clinically stable patient with a suspected leak, CT of the abdomen and pelvis with oral and IV contrast may have higher sensitivity and specificity than UGI contrast studies, with the added utility of identifying 11 ASMBS Position Statements 129 associated intra-abdominal abscesses, hernias, or other not provide traditional durable benefits comparable to curpathologic conditions after GB or SG. Addition of the rently accepted bariatric procedures [18]. chest component to the abdominal CT to rule out disHowever, it has become clear that clinical trials can be, tinct or concomitant pulmonary pathologic conditions and have been, manipulated by for-profit entities, including may be considered. medical device companies, and that this influence on clinical • Given the high mortality from untreated GI leaks, it is trial results can misrepresent the outcomes of the clinical understood that re-exploration, open or laparoscopic, trial. It is therefore essential to ensure the integrity of clinical is an appropriate and acceptable treatment modality trials by recommending the following actions by physicians when a GI leak is suspected and remains the diagnostic involved in clinical trials, as recommended by DeAngelis test with the highest sensitivity and specificity after and Fontanarosa [19] and supported in full by the ASMBS in GB and SG. this Position Statement: “For-profit companies that sponsor 5. Operative management (open or laparoscopic) for acute biomedical research studies should not be solely or primarily GI leaks after GB or SG follows the goals of drainage, involved in collecting and monitoring of data, in conducting placement of drains to create controlled fistulas, use of the data analysis, and in preparing the manuscript reporting antimicrobial agents, and nutrition considerations. study results. These responsibilities should primarily or • Chronic fistulas are described after SG with long clo- solely be performed by academic investigators who are not sure times (≥1–3 months). Definitive surgical manage- employed by the company sponsoring the research.” ment of nonhealing fistulas is technically challenging, Furthermore, the society supports the registration of all cliniand current available data do not favor one procedure cal research trials and mandatory reporting of outcomes, over another. whether favorable or not. 6. Nonoperative management may be an appropriate treatment option for GI leaks after GB or SG in stable patients. • Nonoperative methods of GI leak treatment after both Intragastric Balloon Therapy Endorsed by GB and SG include endoscopic endoluminal self-­ the Society of American Gastrointestinal expandable stents, clips, endoscopic and percutane- and Endoscopic Surgeons ously placed drains, and biologic glue/tissue sealants. Multiple endoscopies and multimodality treatments 1. Level 1 data regarding the clinical utility, efficacy, and may be required to achieve full healing of a chronic safety of intragastric balloon therapy for obesity are fistula. The available data do not favor one treatment derived from randomized clinical studies [20]. over another. 2. Implantation of intragastric balloons can result in notable weight loss during treatment. A few studies, representing lower-level evidence, have suggested that the weight loss Endosurgical Intervention effect can be maintained after balloon retrieval for some for Treatment of Obesity finite time into the future. 3. Although utilization of intragastric balloons results in Currently, a number of endoluminal innovations and novel notable weight loss, separating the effect of the balloon devices and technologies are in various stages of developalone from those of supervised diet and lifestyle changes ment or application to the elective treatment of obesity, may be challenging. Of note, recent FDA pivotal trials including revisional interventions. The theoretical goals of demonstrated a benefit to balloon use compared with diet these therapies include decreasing the invasiveness, risk, and alone in their study populations. In general, any obesity barriers to acceptance of effective treatment of obesity; howtreatment, including intragastric balloon therapy, would ever, these outcomes cannot be assumed and must be proven. benefit from a multidisciplinary team that is skilled and Therefore, the use of novel technologies should be limited to experienced in providing in-person medical, nutritional, clinical trials done in accordance with the ethical guidelines psychological, and exercise counseling. of the ASMBS and designed to evaluate the risk and efficacy 4. The safety profiles for intragastric balloons indicate a safe of the intervention. The results of appropriate trials should intervention, with serious complications being rare. Early include the generation of data for risk-benefit analysis, postoperative tolerance challenges can be significant but assessments of disability, durability, and the resource use can be controlled with pharmacotherapy in the majority associated with the intervention. An intervention undergoing of patients, thereby minimizing voluntary balloon removevaluation should not be judged favorably if the risk/benefit als. These early symptoms should be discussed with the ratio is increased compared with the spectrum of currently patient before the procedure. accepted surgical procedures [17]. A dramatic reduction in 5. Although therapy with prolonged balloon in situ time and risk might allow for the acceptance of interventions that do the use of sequential treatments with multiple balloons 130 have been studied, awareness and adherence to absolute and relative contraindications of use and timely removal optimize device safety. Based on current evidence, balloon therapy is FDA approved as an endoscopic, temporary (maximum 6 months) tool for the management of obesity. Further review will evaluate the impact of diet, lifestyle changes, and pharmacotherapy during and after balloon removal. 6. The ability to perform appropriate follow-up is essential when intragastric balloons are used for weight loss to enhance their safety and avoid complications related to spontaneous deflation and bowel obstruction. Vagal Blocking Therapy for Obesity The American Society for Metabolic and Bariatric Surgery currently supports the following regarding vagal blocking therapy for obesity (VBLOC) for the treatment of obesity and encourages members to participate in post-FDA approval studies [21]: 1. Reversible vagal nerve blockade has been shown to result in statistically significant EWL at 1 year compared with a control group in one of two prospective randomized trials. 2. Reversible vagal nerve blockage has been shown to have a reasonable safety profile with a low incidence of severe adverse events and a low revisional rate in the short term. More studies are needed to determine long-term reoperation and explantation rates. 3. The prospective collection of VBLOC outcomes as part of the national center of excellence databases is encouraged to establish the long-term efficacy of this new technology. ostprandial Hyperinsulinemic Hypoglycemia P After Bariatric Surgery Based on the available evidence to date, the following recommendations are made in the patient with postprandial hyperinsulinemic hypoglycemia after bariatric surgery [22]. 1. Postprandial hyperinsulinemic hypoglycemia after bariatric surgery is rare and most commonly associated with RYGB. Nonetheless, patients should be screened for, educated, and counseled to recognize the signs and symptoms of hypoglycemia. 2. Extreme, progressive, and unrecognized neuroglycopenic symptoms of postprandial hyperinsulinemic hypoglycemia can result in cognitive and neurologic impairment with risk of seizures and loss of consciousness posing risk to both patient and others. S. A. Brethauer and X. (C.) Feng 3. Insulinoma must be ruled out in patients with confirmed fasting hypoglycemia. 4. Diagnosis of postprandial hyperinsulinemic hypoglycemia requires a dietary journal, along with confirmatory laboratory and provocative testing, in the setting of symptoms presenting more than 1 year after surgery. Treatment with dietary modification in mild cases is often implemented successfully without a definitive diagnosis. 5. Postprandial hyperinsulinemic hypoglycemia can be effectively treated in the majority of cases with dietary modification alone. A dietitian should be an integral part of the treatment team and an endocrinologist consulted in cases not responding to initial treatment. 6. Pharmacotherapy produces variable results but should be attempted before surgical intervention. A gastrostomy tube with feeding into the remnant stomach provides nutritional support and in some cases symptomatic relief and should be considered in patients not responding to nonoperative treatment. Partial pancreatectomy is not recommended. Metabolic Bone Changes After Surgery Obesity appears to be independently associated with vitamin and mineral deficiencies involved in bone homeostasis affected by race and potentially affected by gender. These pre-existing vitamin and mineral deficiencies may compound postoperative absorption of bone homeostatic micronutrients depending on the type of weight loss surgery and degree of weight loss. Patients preparing for bariatric surgery should be screened for the presence of vitamin D deficiency and hyperparathyroidism with treatment initiated [23]. Cross-sectional, retrospective, and prospective studies do not conclusively support any increased incidence of osteoporosis or increased fracture risk after bariatric surgery. Accuracy of current methods of assessing bone mineral density (BMD) (DXA) in patients who have extreme obesity as well as after extreme weight loss should be evaluated with further research. The use of one third distal forearm to measure BMD can be considered in situations of extreme weight that exceeds the limits of conventional DXA tables as well as in cases of secondary hyperparathyroidism related to malabsorption of vitamin D and calcium. The degree of bone turnover and BMD loss after bariatric surgery is related to the type of procedure performed, the amount and rate of weight loss, and the degree of malabsorption of other micronutrients and protein. Long-term followup monitoring and supplementation should be provided ­ according the type of procedure and the individual patient’s risk for bone loss. 11 ASMBS Position Statements 1. Preoperative assessment: (a) The high prevalence of vitamin D deficiency and secondary hyperparathyroidism in the obese population supports routine laboratory testing of 25-OHD and intact PTH levels before bariatric surgery, with initiation of treatment for deficiencies and documentation of improvement before surgery when possible. (b) Preoperative DXA can be performed in estrogen-­ deficient women and in premenopausal women and men who have conditions associated with bone loss or low bone mass to establish a baseline before bariatric surgery. There is, however, no compelling data to support routine DXA for all obese adolescents, men or premenopausal women undergoing bariatric surgery. If low bone mass is diagnosed preoperatively, a thorough evaluation should be undertaken to identify secondary causes. This laboratory testing can include thyroid-stimulating hormone and testosterone levels in men. (c) A baseline DXA is recommended by the National Osteoporosis Foundation 2013 (http://www.nof.org/ hcp/practice/practice-and-clinical-guidelines/cliniciansguide) for all women 65 years and older and for younger postmenopausal women and men 70 years or older and men age 50–69 about whom you have concern based on their clinical risk factor profile patients such as those undergoing a malabsorptive procedure. (d) The US Preventative Services Task Force recommends a bone density test at least once for all women age 65 and older. This recommendation can be made in consideration with general medical optimization as indicated. 2. Procedure-specific recommendations for monitoring bone loss are made below. The doses of recommended calcium supplementation and vitamin D supplementation are consistent with previously published 2013 AACE/ TOS/ASMBS Guidelines for the Perioperative Nutritional, Metabolic, and Nonsurgical Support of the Bariatric Surgery Patient [1, 24]. (a) LAGB: • Calcium supplementation after LAGB should include 1200–1500 mg/d, which can be taken in two to three split doses, 4–5 h apart for optimal absorption [25]. In the early postoperative period, minimum vitamin D of at least 3000 IU/d, titrate to 430 ng/mL. Minimum vitamin D maintenance after LAGB should be consistent with established age-specific recommendations for patients at risk for vitamin D deficiency until non-­ obese. The Endocrine Society Clinical Practice Guideline currently recommends a minimum of 600 IU/d of vitamin D from age 19 to 70 years and 800 IU/d of vitamin D after 70þ years [24, 26]. 131 • Bone loss monitoring should include annual albumin, calcium, PTH, and 25-OHD levels. • DXA should be used after LAGB according to the most recent established guidelines for the general patient population that a particular patient belongs to, based on age, gender, and associated risk factors. (b) Gastric bypass and malabsorptive procedures (BPD and BPD/DS): • Supplementation after gastric bypass should include calcium citrate 1200–1500 mg/d, which can be taken in two to three split doses, 4–5 h apart for optimal absorption [27]. Minimum vitamin D intake of 3000 IU/d, titrate to 430 nl/ mL. Calcium citrate is preferable to calcium carbonate due to better ­absorption in the absence or reduction of gastric acid. Supplementation after BPD and BPD/DS should include calcium of 1800–2400 mg/d and minimum vitamin D 3000 IU/d, titrate to 430 nl/mL [1, 24]. • Based on the most recent Endocrine Society Clinical Practice Guidelines, vitamin D deficiencies should be treated with 50,000 IU of vitamin D2 or vitamin D3 once a week for 8 weeks or its equivalent of 6000 IU of vitamin D2 or vitamin D3, daily to achieve a blood level of 25 (OH)D above 30 ng/mL, followed by maintenance therapy of 1500–2000 IU/d. Severe deficiencies can be treated with higher doses up to 50,000 IU three times a day. Intramuscular injections of ergocalciferol 100,000 IU once a week can be used but are rarely necessary [26]. • Bone loss monitoring should include a minimum of annual albumin, calcium, PTH, and 25-OHD levels. 1,25-OH2 D should be monitored in patients with renal compromise. • Bone loss monitoring can also include markers for altered bone turnover. Monitoring to include bone alkaline phosphatase, osteocalcin, serum C-­telopeptide, serum propeptide of type I collagen, and urine N-telopeptides can be considered. The appropriate use of biochemical markers as a screening tool, however, has not been established and warrants additional investigation. • Routine DXA scan after RYGB is not supported by current data. A baseline DXA is recommended by the National Osteoporosis Foundation 2013 (http:// www.nof.org/hcp/practice/practice-and-clinicalguidelines/clinicians-guide) for all women 65 years and older and for younger postmenopausal women and men 70 years and older and men age 50–69 about whom you have concern based on their clinical risk factor profile patients such as those undergoing a malabsorptive procedure. 132 S. A. Brethauer and X. (C.) Feng (c) SG: • Given the current lack of procedure specific data, recommendations regarding supplementation and bone monitoring should at a minimum be consistent with that recommended for the LAGB although it is unlikely that recommendations for the gastric bypass would be harmful. mergency Care of Patients E with Complications After Bariatric Surgery The ASMBS recommends the following guidelines for hospitals and physicians regarding the emergency care of patients with complications related to bariatric surgery procedures [28]: 1. Hospitals are called on to recognize bariatric surgery as a surgical subspecialty, and, similar to the emergency coverage arrangements hospitals provide for other surgical subspecialties, hospitals are required to recognize that a patient with bariatric surgery-related complications should ideally be treated by an appropriately qualified bariatric surgical specialist. Such treatment can be provided by an appropriately qualified and credentialed member of the medical staff in hospitals performing bariatric surgery procedures or by transfer to another facility. Life-threatening conditions that require immediate intervention are appropriately treated by an available general surgeon at hospitals that do not provide bariatric surgery services (see No. 3 below). 2. All hospitals in which elective bariatric surgery procedures are performed are called on to provide care to patients experiencing bariatric surgery-related complications. This is essential to provide a safe system of care, because issues such as relocation, insurance coverage changes, patient access issues, or the nature of an emergency situation can interfere with the provision of care by the primary bariatric surgeon who performed the procedure, their surgical practice, or hospital, as outlined above. Hospitals that provide emergency services to the community and perform bariatric surgical procedures should provide 24-h emergency access to evaluation and treatment (e.g., by way of emergency room coverage) by qualified surgical specialists for all bariatric surgical patients. Hospitals that perform bariatric surgical procedures should also accept the transfer of patients with bariatric surgery-related emergencies from hospitals that do not provide bariatric surgery services. 3. Hospitals that do not perform bariatric surgery might not be equipped to take care of bariatric surgical emergencies and might need to transfer such patients to appropriately equipped centers with qualified bariatric surgical specialists who can treat bariatric surgery-related complications. However, this should only occur if the condition of the patient and specifics of the transfer arrangement will allow safe transfer. Bariatric patients who present with life-threatening surgical problems, whether related to the bariatric surgery or not, should not have their health jeopardized by efforts to arrange such a transfer if it is clear that urgent surgical intervention is indicated. In such circumstances (e.g., closed-loop bowel obstruction threatening perforation or infarction), a general surgeon should perform the lifesaving surgical intervention and avoid the delays inherent in transferring the patient. 4. Bariatric surgeons, as recognized surgical subspecialists, have an obligation to maintain their familiarity with the various bariatric surgical procedures and inherent complications of these procedures as a part of their obligation to provide care to all patients requiring emergency treatment of bariatric surgery-related complications. 5. Bariatric surgeons have an obligation to provide both emergency and elective care to their own postoperative patients, to educate their patients that they are available to provide such care, and to inform their patients how to access this care. 6. Bariatric surgeons must maintain privileges at a hospital with appropriate facilities for bariatric patients that also provide emergency services accessible 24 h daily to care for their bariatric surgery patients who develop complications. Bariatric surgeons who practice exclusively in an outpatient-only facility will not be able to satisfy this obligation and must, therefore, satisfy one of the following criteria: (a) Have in place an approved transfer acceptance emergency care agreement with an appropriate hospital that performs bariatric surgery with emergency services accessible 24 h daily with a qualified bariatric surgeon or surgeons practicing at that facility who will accept their patients and provide emergency care to them. (b) Join the medical staff of an appropriate hospital that performs bariatric surgery and provides emergency services accessible 24 h daily to provide emergency care to their patients who develop bariatric surgery-­ related complications. Global Bariatric Healthcare (Medical Tourism) Based on the limited available data, guidelines published by other medical societies, expert opinion, and a primary concern for patient safety, the American Society for Metabolic and Bariatric Surgery supports the following statements and guidelines regarding bariatric surgical procedures and global bariatric healthcare [29]: 11 ASMBS Position Statements 1. Based on the unique characteristics of the bariatric patient, the potential for major early and late complications after bariatric procedures, the specific follow-up requirements for different bariatric procedures, and the nature of treating the chronic disease of obesity, extensive travel to undergo bariatric surgery should be discouraged unless appropriate follow-up and continuity of care are arranged and transfer of medical information is adequate. 2. The ASMBS opposes mandatory referral across international borders or long distances by insurance companies for patients requesting bariatric surgery if a high-quality bariatric program is available locally. 3. The ASMBS opposes the creation of financial incentives or disincentives by insurance companies or employers that limit patients’ choices of bariatric surgery location or surgical options and, in effect, make medical tourism the only financially viable option for patients. 4. The ASMBS recognizes the right of individuals to pursue medical care at the facility of their choice. Should they choose to undergo bariatric surgery as part of a medical tourism package or pursue bariatric surgery at a facility a long distance from their home, the following guidelines are recommended: • Patients should undergo procedures at an accredited JCI institution or preferably a bariatric center of excellence. • Patients should investigate the surgeon’s credentials to ensure that the surgeon is board eligible or board certified by a national board or credentialing body. Individual surgeon outcomes for the desired procedure should be made available as part of the informed consent process whenever possible. • Patients and their providers should ensure that a follow-­up care, including the management of short- and long-term complications, is covered by the insurance payer or purchased as a supplemental program prior to traveling abroad. • Surgical providers should ensure that all medical records and documentation are provided and returned with the patient to their local area. This includes the type of band placed and any adjustments performed in the case of laparoscopic adjustable gastric banding, as well as any postoperative imaging performed. • Prior to undergoing surgery, the patient should establish a plan for postoperative follow-up with a qualified local bariatric surgery program to monitor for nutritional deficiencies and long-term complications and to provide ongoing medical, psychological, and dietary supervision. • Patients should recognize that prolonged traveling after bariatric surgery may increase the risk of deep venous thrombosis, pulmonary embolism, and other perioperative complications. 133 • Patients should recognize that there are risks of contracting infectious diseases while traveling abroad that are unique to different endemic regions. • Patients should recognize that travel over long distances in a short period of time for bariatric surgery may limit appropriate preoperative education and counseling regarding the risks, benefits, and alternatives for bariatric operations. This also significantly limits the bariatric surgery program’s ability to medically optimize the patient prior to surgery. • Patients should understand that compensation for complications may be difficult or impossible to obtain. • Patients should understand that legal redress for medical errors for procedures performed across international boundaries is difficult. 5. When a patient who has had a bariatric procedure at a distant facility presents with an emergent life-threatening postoperative complication, the local bariatric surgeon on call should provide appropriate care to the patient consistent with the established standard of care and in keeping with ethical guidelines of the ASMBS. This care should be provided without risk of litigation for complications or long-term sequelae resulting from the initial procedure performed abroad. Routine or non-emergent care for patients who have had bariatric surgery elsewhere should be provided at the discretion of the local bariatric surgeon. Question Section 1. The purpose of an ASMBS position statement is to: A. Clarify a controversial issue B. Provide guidance for healthcare leaders and payers C. Provide support for clinical decisions made by the membership D. All of the above 2. The conclusions in the “Preoperative Weight Loss Requirement” position statement include: A. Every patient, regardless of BMI, should undergo at least 6 months of supervised dieting before surgery. B. There is no level 1 evidence to support mandatory preoperative weight loss, and this policy often delays care. C. The policy of mandatory supervised diet policies for every patient is acceptable because bariatric patients need to demonstrate the willpower needed to be successful after surgery. D. The ASMBS supports insurance-mandated preoperative weight loss policies for all patients seeking bariatric surgery. 134 3. Based on current studies, which of the following statements regarding alcohol use after gastric bypass are false? A. RYGB results in accelerated alcohol absorption (shorter time to reach maximum concentration). B. RYGB causes higher maximum alcohol concentration. C. After RYGB, it takes a longer time to eliminate alcohol in men but not in women. D. RYGB poses an increased risk for development of alcohol use disorder. 4. Position statements and guidelines published by the ASMBS: A. Are written by the Clinical Issues Committee, sometimes in collaboration with another ASMBS committee or another professional society B. Undergo a thorough vetting and revision process C. Are sent to the membership for comment before final approval D. Are reviewed every 3 years to determine if updates are needed E. All of the above References 1. Mechanick JI, Youdim A, Jones DB, et al. Clinical practice guidelines for the perioperative nutritional, metabolic, and nonsurgical support of the bariatric surgery patient—2013 update: cosponsored by American Association of Clinical Endocrinologists, the Obesity Society, and American Society for Metabolic & bariatric surgery. Obes Silver Spring Md. 2013;21:S1–27. 2. ASMBS. ASMBS statement for access to care for obesity treatment available from: https://asmbs.org/resources/access-to-care-for-obesity-treatment. 2011. 3. Kim JJ, Rogers AM, Ballem N, et al. ASMBS updated position statement on insurance mandated preoperative weight loss requirements. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2016;12:955–9. 4. Kominiarek MA, Jungheim ES, Hoeger KM, et al. American Society for Metabolic and Bariatric Surgery position statement on the impact of obesity and obesity treatment on fertility and fertility therapy endorsed by the American College of Obstetricians and Gynecologists and the Obesity Society. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2017;13:750–7. 5. Parikh M, Johnson JM, Ballem N, et al. ASMBS position statement on alcohol use before and after bariatric surgery. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2016;12:225–30. 6. Heinberg LJ, Ashton K, Coughlin J. Alcohol and bariatric surgery: review and suggested recommendations for assessment and management. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2012;8:357–63. 7. Aminian A, Chang J, Brethauer SA, et al. ASMBS updated position statement on bariatric surgery in class I obesity (BMI 30–35 kg/m2). Surg Obes Relat Dis Off J Am Soc Bariatr Surg. Epub ahead of print June 9, 2018. DOI: https://doi.org/10.1016/j.soard.2018.05.025. 8. ASMBS Clinical Issues Committee. Peri-operative management of obstructive sleep apnea. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2012;8:e27–32. 9. American Society for Metabolic and Bariatric Surgery Clinical Issues Committee. ASMBS updated position statement on prophylactic measures to reduce the risk of venous thromboembolism S. A. Brethauer and X. (C.) Feng in bariatric surgery patients. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2013;9:493–7. 10. Clinical Issues Committee of American Society for Metabolic and Bariatric Surgery. Sleeve gastrectomy as a bariatric procedure. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2007;3:573–6. 11. Clinical Issues Committee of the American Society for Metabolic and Bariatric Surgery. Updated position statement on sleeve gastrectomy as a bariatric procedure. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2010;6:1–5. 12. ASMBS Clinical Issues Committee. Updated position statement on sleeve gastrectomy as a bariatric procedure. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2012;8:e21–6. 13. Ali M, El Chaar M, Ghiassi S, et al. American Society for Metabolic and Bariatric Surgery updated position statement on sleeve gastrectomy as a bariatric procedure. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2017;13:1652–7. 14. Kim J. American Society for Metabolic and Bariatric Surgery statement on single-anastomosis duodenal switch. Surg Obes Relat Dis. 2016;12:944–5. 15. Clinical Issues Committee. ASMBS policy statement on gastric plication. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2011;7:262. 16. Kim J, Azagury D, Eisenberg D, et al. ASMBS position statement on prevention, detection, and treatment of gastrointestinal leak after gastric bypass and sleeve gastrectomy, including the roles of imaging, surgical exploration, and nonoperative management. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2015;11:739–48. 17. Buchwald H. Consensus conference panel. Consensus conference statement bariatric surgery for morbid obesity: health implications for patients, health professionals, and third-party payers. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2005;1:371–81. 18. Brethauer SA, Pryor AD, Chand B, et al. Endoluminal procedures for bariatric patients: expectations among bariatric surgeons. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2009;5:231–6. 19. DeAngelis CD, Fontanarosa PB. Impugning the integrity of medical science: the adverse effects of industry influence. JAMA. 2008;299:1833–5. 20. Ali MR, Moustarah F, Kim JJ, et al. American Society for Metabolic and Bariatric Surgery position statement on intragastric balloon therapy endorsed by the Society of American Gastrointestinal and Endoscopic Surgeons. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2016;12:462–7. 21. Papasavas P, El Chaar M, Kothari SN, et al. American Society for Metabolic and Bariatric Surgery position statement on vagal blocking therapy for obesity. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2016;12:460–1. 22. Eisenberg D, Azagury DE, Ghiassi S, et al. ASMBS position statement on postprandial hyperinsulinemic hypoglycemia after bariatric surgery. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2017;13:371–8. 23. Kim J, Brethauer S, ASMBS Clinical Issues Committee, et al. Metabolic bone changes after bariatric surgery. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2015;11:406–11. 24. Mechanick JI, Kushner RF, Sugerman HJ, et al. American Association of Clinical Endocrinologists, the Obesity Society, and American Society for Metabolic & bariatric surgery medical guidelines for clinical practice for the perioperative nutritional, metabolic, and nonsurgical support of the bariatric surgery patient. Endocr Pract Off J Am Coll Endocrinol Am Assoc Clin Endocrinol. 2008;14(Suppl 1):1–83. 25. Compston JE, Vedi S, Gianetta E, et al. Bone histomorphometry and vitamin D status after biliopancreatic bypass for obesity. Gastroenterology. 1984;87:350–6. 26. Holick MF, Binkley NC, Bischoff-Ferrari HA, et al. Evaluation, treatment, and prevention of vitamin D deficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2011;96:1911–30. 11 ASMBS Position Statements 27. Heaney RP, Recker RR, Saville PD. Calcium balance and calcium requirements in middle-aged women. Am J Clin Nutr. 1977;30:1603–11. 28. Clinical Issues Committee of the American Society for Metabolic and Bariatric Surgery. American Society for Metabolic and Bariatric Surgery position statement on emergency care of patients 135 with complications related to bariatric surgery. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2010;6:115–7. 29. American Society for Metabolic and Bariatric Surgery Clinical Issues Committee. American Society for Metabolic and Bariatric Surgery position statement on global bariatric healthcare. Surg Obes Relat Dis Off J Am Soc Bariatr Surg. 2011;7:669–71. Part II Primary Bariatric Surgery and Management of Complications Laparoscopic Gastric Bypass: Technique and Outcomes 12 Kelvin D. Higa and Pearl K. Ma Chapter Objectives 1. Discuss the basic construct and operative techniques used in laparoscopic gastric bypass. 2. Discuss patient preparation. 3. Review long-term outcomes. Introduction Historically, the first laparoscopic gastric bypass performed by Wittgrove and Clark in 1994 was a remarkable achievement [1]. Their pioneering work accelerated public acceptance and industry interest in bariatric surgery that led to further innovation and helped define this specialty. Variations of this original technique exist, but the basic tenets of the procedure remain the same: small, isolated gastric pouch, limited diversion of bilioenteric secretions, and reproducible, safe anastomotic methods. During this time period, the vertical banded gastroplasty was declining in popularity, giving way to the open gastric bypass as it evolved into a more standardized operation. The original gastric bypass described by Mason in 1966 [2] has little similarity to operations performed today, so it is unfair to compare earlier outcomes to current procedures. However, it was because of this early experience and subsequent modifications adopted in order to avoid complications that our present laparoscopic procedures owe their lineage. Our current understanding of the physiologic effects of the gastric bypass is as naïve as our understanding of the disease for which we are treating: morbid obesity. Early constructs were based on the anatomic “restrictive” or “malabsorptive” concepts that did not correlate well with our observation of patient behavior. Why was there such a proK. D. Higa (*) · P. K. Ma Department of Surgery, Minimally Invasive and Bariatric Surgery, Fresno Heart and Surgical Hospital, University of California San Francisco, Fresno, CA, USA nounced effect on metabolic syndrome prior to weight loss, and how did our patients maintain “satiety” without a seemingly “restrictive” component? With the discovery and further understanding of enteric hormones, such as gastrin, GLP-1, and PYY, the hypothesized mechanism(s) of action of the gastric bypass fits better with our long-standing clinical observations [3]. The effect on the individual patient, by whatever means, is reproducible—but these proposed mechanisms also explain the variability of the response of each individual as well as consistent response of the group given the wide range of anatomic variability in the anatomic construct. In other words, a pouch that varies in volume of 10 cc compared to one of 30 cc, a 200% difference, has not been shown to increase weight loss or improve outcomes. Likewise, creating a Roux limb of 150 cm does not impart greater effect than one of 75 cm. Although there are studies that have shown short-term benefit of pouch and/or stoma reduction and lengthening of the Roux limb to enhance weight loss, results have been inconsistent and without long-­ term benefit. The best predictor of success seems to be genetic similarity among related individuals, rather than environmental factors [4]. The performance of the gastric bypass may not be influenced as strongly by compliance of the patient as it seems to be with the adjustable gastric band and may be predetermined by the genetic and biologic nature of each individual patient. Operative strategy, therefore, is the same as it was in 1966: to achieve the anatomic effect of proximal alimentary diversion, with the least side effects and complications with long-term control of weight and medical comorbidities— safely and cost-effectively. The laparoscopic gastric bypass was once considered to be one of the most challenging minimally invasive operations. It is now the most common foregut operation and, despite its complexity and learning curve, has been shown to be safer and more cost-effective than its open predecessor. The operation has evolved to include a variety of anasto- © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_12 139 140 motic techniques and trocar placements, giving individual surgeons the latitude to adopt or modify the procedure based on their own preference and experience. This chapter will discuss the basic construct and operative techniques in use today. There is no one method that is the “gold standard.” Comparative studies are few, and those that exist only reinforce that there are subtleties to operative technique that defy observation. For example, gastrojejunal stricture rate may be influenced more by preservation of blood supply and operative technique rather than the diameter of the circular stapler. It would be naïve to expect every surgeon to achieve the same results, given the same basic technique; it is up to each individual to modify, adapt, and improve through his or her own experience. K. D. Higa and P. K. Ma Trocar placement is a highly variable and yet critical step toward a safe and successful operation. Most authors describe external landmarks such as the umbilicus or xiphoid to determine placement. However, obese patients have a high degree of abdominal wall thickness with corresponding varying degrees of rigidity. Also, the size of the liver and presence of previous operations and their associated internal adhesions will determine initial and subsequent trocar placement. One must recognize that placement of the trocars needs to accommodate manipulation and construction of both the small bowel and hiatus, often challenging with the larger patients. Therefore, we feel it better to place the trocars based on internal anatomy, rather than external landmarks. In this way, triangulation and visualization will be preserved, accommodating for variations in the size of the liver or length of the patient’s torso. Preparation of the Patient Attention must be given not only to individual trocar placement but also to the angle in which the trocar enters the Psychosocial and nutritional education is paramount to long-­ skin. Some individuals’ thick, muscular abdominal wall does term success with any bariatric/metabolic operation and is not allow for the range of motion necessary to achieve the covered in detail in other chapters and in volume 2 (The objective, forcing redirection of the trocar internally, through ASMBS Textbook of Bariatric Surgery, Volume II: Integrated the same skin incision, but different fascial opening, or by Health). In general, most patients fail to appreciate the life- placement of another trocar. In general, the optimal placestyle changes experienced after surgery despite a high degree ment is to orient all trocars toward the midline, pointing to of preparation; postoperative support and education are nec- the base of the mesocolon. Extra-long trocars may be necessary. Although some suressary requirements and should be made available lifelong. Concurrent health-care maintenance with evaluation and geons prefer to limit the number of 12 mm trocars (necessary optimization of cardiovascular and pulmonary risks is appro- to accommodate stapling devices), this may limit proper stapriate for all elective procedures. Mandatory preoperative pler orientation and compromise the anatomic construct. The weight loss can be effective in decreasing the size of the liver hernia risk is minimized by either closing the trocar defects but has not been shown to improve outcomes or diminish or, preferably, using non-bladed trocars without fascial closure. rates of complication or open conversion. An example of a trocar placement scheme is as follows: Thromboembolism prophylaxis and perioperative antibiits purpose is only to illustrate the rationale necessary for otics are currently the standard of care. consistency of this important and underappreciated first step in performing the laparoscopic gastric bypass. Variations, depending on experience, technique, and judgment, are necPositioning and Trocar Placement essary for evolution to occur and should be encouraged: Positioning of the patient is by surgeon preference and should take into consideration patient habitus and avoidance 1. Initial trocar (12 mm)—left, upper quadrant, subcostal, of pressure points (Fig. 12.1). Many surgeons prefer the and midclavicular line. This is often an optical entry lithotomy or “French” position, standing between the without prior insufflation. The rationale is that many patient’s legs in steep reverse Trendelenburg, while others patients have had previous procedures; pelvic or otherwill operate from the side, with the legs together. Either wise—this area is rarely affected with intra-abdominal way, one must consider the weight capacity of the operating adhesions from common open procedures. This allows table along with adequate securing measures when the need dissection of midline adhesions, inspection of the size for lateral extension in extreme cases should arise. One must of the liver, and determination of the best level for the also consider ergonomics of the operative surgeon and assisprimary optical port. This will also be the primary port tant personnel. The surgeon and supportive staff are obliged for vertical stapling of the gastric pouch. Once adheto operate in a safe and comfortable environment so as to sions are mobilized, then the optical port can be avoid back and neck injury or carpal tunnel syndrome from thoughtfully placed as to see the ligament of Treitz as suboptimal positioning or unavoidable repetitive motion. well as hiatus without having to “turn around.” Also, by 12 Laparoscopic Gastric Bypass: Technique and Outcomes 141 Fig. 12.1 Operating room arrangement and port placement keeping the initial entry away from the midline, the vena cava and aorta are not as vulnerable to injury. 2. Primary optical trocar (12 mm)—placement has been described above. Optimal placement allows for forward visualization of the proximal small bowel and the hiatus. Once this trocar is placed, the camera is moved to this port for subsequent trocar placement. I have not found the current 5 mm scopes to provide enough light delivery and therefore resolution for optimal visualization in most patients. 3. Right-sided trocar (12 mm)—this trocar must be placed thoughtfully just as all others. Exterior landmarks are irrelevant. It must come in below the liver edge, just to the right of the midline so as to be able to triangulate on the hiatus as well as the ligament of Treitz; therefore, it should be angled toward the root of the mesocolon, rather than perpendicular to the abdominal wall. It must be 12 mm to accommodate the stapler that will define the inferior gastric pouch. 4. Left inferior trocar (12 mm)—this is often at the same level as the primary optical trocar and in the same line as the initial trocar. This will be the primary stapler entry site for the jejunojejunostomy (Fig. 12.2) and along with the Fig. 12.2 Steps in jejunojejunostomy 142 Fig. 12.3 Steps in gastrojejunostomy right upper quadrant trocar will triangulate very well for a comfortable manual gastrojejunostomy (Fig. 12.3). 5. Liver retractor—the most consistent placement appears to be the subxiphoid. A 5 mm trocar can be used here, depending on the liver retractor of choice. We have found that a simple 5 mm instrument or similar device will provide excellent exposure and therefore is often placed without a trocar, through direct puncture, as it will not be removed until the end of the case. The Components The Pouch Much emphasis is placed on the formation of the gastric pouch or modification of the gastric pouch to improve performance. Its contribution to the “gastric bypass” effect is undeniable, yet poorly understood. Collateral evidence suggests that the actual size and configuration of the pouch are more important for prevention of complications and improved alimentation rather than maximal weight loss or metabolic effect. In other words, one would be hard-pressed to find literature, anecdotal, or otherwise that describes a linear correlation between pouch size and weight loss. Likewise, attempts at pouch reduction for weight recidivism or inadequate weight loss have been inconsistent and disappointing. Early gastric bypass descriptions were of horizontal orientations and often nondivided staple lines. Both have been retired with the adoption of linear cutting stapling devices K. D. Higa and P. K. Ma allowing for orientation and construct based on intent, rather than necessity. One can now orient the pouch vertically with complete isolation of the gastric pouch with more precision and less concern for splenic or other visceral injury. This was made even easier with the laparoscopic stapling devices; along with the increased visualization of the proximal stomach and hiatus, the laparoscopic approach has improved the safety and reproducibility of this procedure. Isolation of the gastric pouch was important to limit the potential of gastrogastric fistula—relatively common complication of nondivided staple lines. It also allowed for more precise dissection—nondivided pouches were often created by placement of a spherical intragastric balloon for sizing, rather than a cylindrical bougie. This made the pouch consist primarily of anterior stomach wall with a greater amount of fundus that accounted for its eventual dilation and increased capacity. Vertical, lesser curve-based orientation of the pouch has been shown by Mason to be ideal for gastroplasty, avoiding the distensible fundus, making the pouch more stable in size, an observation. The pouch is formed by sequential firing of a laparoscopic linear cutter stapler, a stapling device around an intraluminal bougie. The first firing is usually horizontal beginning no more than 5 cm distal to the esophagogastric junction; subsequent firings are vertically oriented to the angle of His. Creating a longer pouch may lead to an increased risk of developing marginal ulcerations in the future [5]. The staple height depends upon the thickness of the tissue, often requiring 3.8–4.1 mm cartridges. The size of the bougie does not appear to be controversial; most surgeons use the same size to calibrate the gastrojejunal anastomosis—usually 1.2– 1.5 mm diameter. One of the critical steps in creation of the gastric pouch is posterior visualization at the level of the hiatus. Many surgeons will “bluntly” dissect behind the stomach, but given the variable level of adhesions to the pancreas and the splenic vessels, this is unwise. Optimally, it is better to enter the lesser sac through the gastrocolic omentum and free the posterior gastric adhesions up to the esophageal hiatus. This protects the pancreas and the occasional tortuous splenic artery from inadvertent injury. After this, the lesser curve, perigastric dissection can be performed with more confidence and placement of the stapler more precisely so as not to “twist” the stomach pouch. This occurs when posterior gastric adhesions prevent the initial horizontal stapler from capturing equal amounts of anterior and posterior gastric wall. The resultant twist is not as critical as in the gastric sleeve but looks less than ideal nonetheless. More controversial is whether or not to dissect the hiatus and repair a hiatal hernia when present. Autopsy studies show that a hiatal hernia is present in up to 70% of individuals, similar to our observations when we routinely dissect out the hiatus in all patients. However, dissection of the hiatus 12 Laparoscopic Gastric Bypass: Technique and Outcomes can add additional time and potential complications to an already complicated procedure. Our studies have not shown that preoperative endoscopy accurately predicts the absence of a hiatal hernia; the only way to determine its presence is circumferential dissection of the esophagus. The absence of the “anterior” dimple is not reliable as the hernia space is often taken up by a large paraesophageal lipoma that can be easily reduced into the abdomen once identified. Once identified, the hiatal hernia is best repaired posteriorly with permanent suture. The question remains: “Is it important to repair every hiatal hernia at the time of gastric bypass?” The answer is not clear. If one assumes that precise dissection and formation of the gastric pouch are important to limit postoperative complications, then it would be appropriate to absolutely identify the location of the gastroesophageal junction—often hidden in a “sea of fat”—to better perform more consistent reconstruction. It has been our observation that almost all patients undergoing revision after gastric bypass have a significant hiatal hernia at the time of reoperation—something not appreciated at the time of the first intervention. Dissection of the hiatus and repair of the hiatal hernia along with removing the fat pad overlying the angle of His may allow for more precise and consistent pouch formation and subsequent better long-term performance and lower complications. However, this has not been proven. In addition, disruption of the phreno-esophageal ligament may predispose the patient to recurrent paraesophageal herniation, thus complicating their postoperative course. Postoperative GERD does occur after gastric bypass, but probably not attributable to an undiagnosed hiatal hernia as much as it is because of a poorly formed pouch and redundant fundus. The Bypass The vertical banded gastroplasty has not been shown to be as effective as the gastric bypass in terms of long-term weight loss and quality of life owing to significant GERD and persistence of appetite and hunger. Clearly, the intestinal bypass is an integral part of the physiology of the gastric bypass even though the mechanism of its contribution is still largely unknown. Overall intestinal length can vary as much as 100%, and intraoperative measurements are far from precise given the dynamism of the small bowel. Still, attempts to accurately measure and modify gastric bypass limb lengths to correlate with weight loss and malnutrition have been published. Very few comparative studies exist; most show no difference except in the super obese population and then only for a few years. Our data suggests no difference between a 100 and 150 cm Roux limb when stratified to patients with a BMI greater or lower than 50 kg/m2. Varying biliopancreatic limb 143 lengths up to 100 cm likewise have failed to show a difference. Therefore, it does not seem critical or necessary to expose gastric bypass patients to excessive diversion beyond that which is necessary to prevent bile reflux to the gastric pouch. Extreme variants of the proximal gastric bypass that include radically lengthening the Roux limb, shortening the common channel to 50–150 cm from the ileocecal valve (type: 2 distal gastric bypass), or shortening the entire alimentary limb length to 3–4 m (type: 1 distal gastric bypass), effectively creating a biliopancreatic-type diversion, do not constitute what is commonly referred to by patients and insurers as “gastric bypass.” The length of the biliopancreatic limb is not critical, usually just long enough to provide mobilization of the Roux limb to the gastric pouch without tension. This depends on whether or not the Roux limb is to be routed antecolic or retrocolic, with antecolic naturally requiring more length. The length of the Roux limb should be at least 75 cm as we have seen bile reflux in patients with 60 cm Roux limbs. Variants up to 150 cm give no added benefits. Beyond 150 cm, insufficient data exists to illuminate any benefit. The bowel is transected with a linear cutting stapling device. Surprisingly, little mesentery division needs to be performed; one must be careful not to transect the superior mesenteric artery. Construction of the jejunojejunostomy is usually performed as a side-to-side anastomosis with linear cutter staplers (Fig. 12.2). Attention must be directed to avoid kinking or twisting the Roux limb or inadvertently performing the notorious Roux-en-O by not properly identifying each limb prior to anastomosis. The Anastomosis The least controversial but most often studied component of the laparoscopic gastric bypass is the gastrojejunal anastomosis. Three distinct methods exist. The original method proposed by Wittgrove and Clark involved endoscopic retrieval of a percutaneous guidewire that was then attached to the anvil of a 21 mm circular stapler. This allowed the anvil to be passed orally into the gastric pouch; the stapler was passed through an enlarged port site through an opening of the Roux limb so as to create the anastomosis. After retrieval, the afferent limb was then transected. The anastomosis can then be inspected and tested with the flexible endoscope. Care must be taken when the anvil passes the cricopharyngeus—the narrowest part of the esophagus. Scott and de la Torre modified placement of the anvil through a gastrotomy prior to gastric pouch formation, elimi- 144 K. D. Higa and P. K. Ma nating the need for operative endoscopy or transoral passage of the anvil [6]. The gastrotomy required closure similar to the enterotomy made on the Roux limb with the Wittgrove method. Other surgeons used the linear cutter stapler to create a side-to-side anastomosis from the Roux limb to the gastric pouch [7]. This technique required manual closure or the residual opening, but minimal suturing was required. The simplest but overlooked method of creating this anastomosis is a handsewn approach familiar to most open bariatric surgeons. It is unclear why many minimally invasive surgeons dismiss this technique as being too difficult when much of the operation still requires manual suturing skill. The handsewn anastomosis remains the most cost-effective method of gastrojejunostomy. It remains the surgeon’s choice as to the absorbable suture material, single or two layers, and interrupted or continuous, as well as the diameter of the thread and size of the needle. We prefer 3–0 absorbable sutures for this application. outing of the Roux Limb and Closure R of Mesenteric Defects The Roux limb can be brought through the mesocolon (retrocolic) or anterior to the colon (antecolic) as well as anterior or posterior to the gastric remnant. While all routes are acceptable, one must be familiar with all methods so as to be able to adapt to any situation. For example, if one has planned, or the situation requires, a gastrostomy tube, then a retro-gastric placement of the Roux limb will allow the gastric remnant to attach, unimpeded, to the abdominal wall. The antecolic routing eliminates one potential site of herniation—the mesocolon—but introduces additional tension on the gastrojejunal anastomosis by the weight of the colon and undivided omentum, if present. When necessary, the omentum can be shifted to the right of the patient or divided; under no circumstance is a trans-omental route acceptable for the potential for herniation through the omental defect and possible small bowel obstruction. The large Petersen’s space should be closed either way. There is no clear advantage of routing the bowel behind or in front of the colon. If the mesocolon were excessively short, it would be prudent to go antecolic. If the omentum is heavy or difficult to manipulate or a short small bowel mesentery is found, a retrocolic route would be more efficient. When the omentum is adherent to the pelvis or lower abdomen, as in the case of previous surgery, a supra-mesocolic approach to the small bowel can be most efficient. The retrocolic route for the Roux limb leaves three potential sites of internal herniation: the jejunojejunostomy space, the mesocolon, and the Petersen’s space. With continuous closure with nonabsorbable sutures, the internal hernia rate should approach 1%. Although there are some advocates for not closing potential sites of herniation (citing personal experience), given the potential serious and emergent nature of small bowel obstruction after gastric bypass from internal herniation and the minimal risk of complications from mesenteric closure, leaving these spaces open makes little sense [8]. Outcomes Long-term data regarding gastric bypass have been lacking due to the complexity of issues regarding follow-up [9–14]. Himpens [14] reported 9-year data consistent with long-term open gastric bypass data that was comparable to our 10-year data [13]. Although we experienced poor follow-up, there was no difference in outcomes between patients who followed up in our office and those who did not; so it is not unreasonable to extrapolate our results. Consistent with most series (Table 12.1; Fig. 12.4), there was gradual weight gain over time, stratified to the severity of obesity; individuals with BMI < 50 kg/m2 were more likely to lose a greater percentage of their excess weight initially but tended to regain weight similar to the super obese. Table 12.1 Long-term postoperative BMI Investigator Jones [9] Pories et al. [10] Sugerman et al. [11] Christou et al. [12] Higa et al. [13] Himpens et al. [14] Patients (n) 352 608 1025 Follow-up (years) 10 10 10–12 Patients eligible for follow-up Patients at follow-up, n (n) (%) 71 36 (51) NR 158 (NR) 361 135 (37) % EWL 62 55 52 Postoperative BMI (kg/ m2) 30 35 36 272 12 272 161 (59) 68 38 242 126 10 9 242 126 65 (27) 77 (61) 57 63 33 30 12 Laparoscopic Gastric Bypass: Technique and Outcomes Fig. 12.4 % Excess weight loss for patients who had preoperative BMI < 50 kg/m2 compared to those with preoperative BMI ≥ 50 kg/m2 [13] 145 preBMI <50 kg/m2 (143 pts) preBMI ≥50 kg/m2 (53 pts) % Excess Weight Loss 70 60 50 0 2 4 6 Postoperative Years 8 10 Table 12.2 Postoperative comorbid conditions for patients in Higa et al.’s study [13] Outcomes of comorbid conditions for 242 study patients and 51 patients evaluated during postoperative year 10 242 study patients 51 patients with 10-year follow-up Patients % of Comorbid condition (n) 242 Follow-up (%) Resolved or improved (%) Follow-up (%) Resolved or improved (%) Osteoarthritis 110 45 35 84 100 78 Diabetes 45 19 27 83 75 67 Dyslipidemia 6 2 100 67 100 80 Hypertension 108 45 36 87 100 86 Infertility 5 2 40 50 100 100 Obstructive sleep apnea 45 19 47 76 95 79 Asthma 23 10 30 100 100 100 Gastroesophageal reflux disease 121 50 36 89 94 90 Urinary stress incontinence 35 14 46 69 92 55 Varicose veins 21 9 29 100 63 100 Resolution of comorbid conditions was significant, but there is a trend for recurrence of diabetes over time (Table 12.2) [13]. This is consistent with other reports. Adams [15] showed a reduction of remission of diabetes after gastric bypass from 75% to 62% from years 2–6 post­op, while Himpens reported no recidivism of diabetes after 9 years but, ironically, a 27.9% incidence of new-onset diabetes. Resolution and/or improvement of dyslipidemia, hypertension, asthma, GERD, and obstructive sleep apnea appears to be sustainable. Despite some weight regain over time, reduction in overall mortality has been observed [16–18]. Mortality rates now approach 0.16% [19] primarily due to pulmonary thromboembolism with anastomotic or stapler leak rates stabilizing at 1%. Early complications (Tables 12.3 and 12.4) are primarily due to gastrojejunal stenosis, amenable to endoscopic balloon dilation. Late complications include marginal ulceration, biliary tract disease, internal hernia, and alcohol dependency [20, 21]. Table 12.3 Complications among postoperative patients in Higa et al.’s study [13] Complications of 242 study patients and 65 patients evaluated during postoperative year 10 65 patients evaluated 242 study during POY 10, n patients, n (%) (%) Morbidity Early Incomplete division of 4 (1.7) stomach Staple line failure 2 (0.8) Thermal injury with 1 (0.4) perforation Marginal ulcer perforation 1 (0.4) Bleeding, observation 1 (0.4) 1 (1.5) Deep venous thrombosis 1 (0.4) Stenosis, gastrojejunostomy 12 (5.0) 4 (6.2) Stenosis, mesocolon 1 (0.4) Fever 1 (0.4) Readmission 5 (2.1) 1 (1.5) Hypoglycemia 1 (0.4) 1 (1.5) (continued) 146 Table 12.3 (continued) Complications of 242 study patients and 65 patients evaluated during postoperative year 10 65 patients evaluated 242 study during POY 10, n patients, n (%) (%) Morbidity Central pontine myelinolysis 1 (0.4) Subtotal 31 (12.8) 7 (10.8) Late Internal hernia 39 (16.1) Marginal ulcer 11 (4.5) 5 (7.7) Gastrogastric fistula 1 (0.4) 1 (1.5) Biliary requiring 17 (7.0) 12 (18.5) cholecystectomy Alcohol dependency 6 (2.5) 5 (7.7) Other substance abuse 1 (0.4) Hernia, trocar 3 (1.2) 1 (1.5) Subtotal 78 (32.2) 24 (36.9) Total 109 (45.0) 31 (47.7) Data in parentheses are percentages POY postoperative year Table 12.4 Postoperative nutritional deficiencies for 136 patients over 10 years Cumulative incidence of nutritional deficiencies for 136 study patients tested between postoperative years 1 and 10 Patients with Percent of 136 Parameter deficiency (n) patients tested 81 60 Vitamin B12 Hemoglobin 71 52 Albumin 46 34 Intact parathyroid hormone 36 26 Vitamin B6 23 17 Calcium 18 13 Folate 2 1 Conclusion The laparoscopic gastric bypass has proven to be superior to the traditional open approach with respect to complications, cost, patients’ acceptance, and possibly weight maintenance. In addition, the minimally invasive approach and current instrumentation have allowed for refinement, improved precision, and standardization of the procedure through video documentation and peer review criticism of operative technique not possible with open surgery. Clearly, the laparoscopic approach can now be considered the standard of care, similar to cholecystectomy—no one should be offered an open operation in the elective setting. However, the learning curve for this operation is long, even with good laparoscopic skills and intense mentorship as in the case of most MIS fellowship programs. Given the low incidence of serious complications with the associated expectations of superior outcomes and the need for a supportive program, it is not reasonable that surgeons can or K. D. Higa and P. K. Ma should perform this operation without fellowship training or mentorship from senior partners. Although the pathophysiology of the gastric bypass is still debated, the outcomes, its effect on metabolic syndrome, and its underutilization cannot be denied. In the absence of evidence-­based data, surgeons are forced to rely upon anecdotal and observational data when constructing or modifying their procedures. Given the seemingly infinite variables that can contribute to results, including reliance upon the compliance of patients themselves, observational data may be more applicable in a given practice than the illusion of randomized trials when comparing nuances in operative technique. Further modifications such as robotic applications and single-incision surgery have yet to define superior results; their true advantages, that of marketing, may have relevance in certain demographics, but as surgeons we must be cautious when defining our role in health care. Question Section 1. What was the original anastomotic technique described by Wittgrove and colleagues for construction of the gastrojejunal anastomosis? A. Handsewn B. Linear stapler C. Circular stapler 2. What is the most common cause of death after gastric bypass? A. Obstruction B. Bleeding C. Sepsis D. Pulmonary embolism 3. What step is critical in creating the gastric pouch? A. Posterior visualization at the level of the hiatus B. Horizontal-based pouch to include the fundus of stomach C. Disruption of the phreno-esophageal ligament D. Pouch size greater than 7 cm 4. Internal herniation can occur in which potential space(s) after creation of retrocolic gastric bypass? A. Petersen’s space B. Jejunojejunostomy space C. Transverse mesocolon space D. All of the above References 1. Wittgrove AC, Clark GW, Tremblay LJ. Laparoscopic gastric bypass, Roux-en-Y: preliminary report of five cases. Obes Surg. 1994;4:4353–7. 2. Mason EE, Ito C. Gastric bypass in obesity. Surg Clin North Am. 1967;47:1345–51. 12 Laparoscopic Gastric Bypass: Technique and Outcomes 3. Evans S, Pamuklar Z, Rosko J, Mahaney P, Jiang N, Park C, Torquati A. Gastric bypass surgery restores meal stimulation of the anorexigenic gut hormones glucagon-like peptide-1 and peptide YY independently of caloric restriction. Surg Endosc. 2012;26(4):1086–94. 4. Hatoum IJ, Greenawalt DM, Cotsapas C, Reitman ML, Daly MJ, Kaplan LM. Heritability of the weight loss response to gastric bypass surgery. J Clin Endocrinol Metab. 2011;96(10):1630–3. 5. Ehdolm, et al. Importance of pouch size in laparoscopic Roux-en-Y gastric bypass: a cohort study of 14,168 patients. Surg Endosc. 2016;30:2011–5. 6. De la Torre RA, Scott JS. Laparoscopic Roux-en-Y gastric bypass: a totally intra-abdominal approach—technique and preliminary report. Obes Surg. 1999;9:101–6. 7. Williams MD, Champion JK. Linear technique of laparoscopic Roux-en-Y gastric bypass. Surg Technol Int. 2004;13:101–5. 8. Iannelli A, Facchiano E, Gugenheim J. Internal hernia after laparoscopic Roux-en-Y gastric bypass for morbid obesity. Obes Surg. 2006;16:1265–71. 9. Jones K. Experience with the Roux-en-Y gastric bypass, and commentary on current trends. Obes Surg. 2000;10:183–5. 10. Pories W, Swanson M, MacDonald K. Who would have thought it? An operation proves to be the most effective therapy for adult-onset diabetes mellitus. Ann Surg. 1995;222:339–50. 11. Sugerman HJ, Wolfe LG, Sica DA, Clore JN. Diabetes and hypertension in severe obesity and effects of gastric bypass-induced weight loss. Ann Surg. 2003;237:751–8. 12. Christou NV, Look D, Maclean LD. Weight gain after short- and long-limb gastric bypass in patients followed for longer than 10 years. Ann Surg. 2006;244:734–40. 147 13. Higa KD, Ho T, Tercero F, Yunus T, Boone KB. Laparoscopic Roux-en-Y gastric bypass: 10-year follow-up. Surg Obes Relat Dis. 2011;7:516–25. 14. Himpens J, Verbrugghe A, Cadiėre G, Everaerts W, Greve J. Long-­ term results of laparoscopic Roux-en-Y gastric bypass: evaluation after 9 years. Obes Surg. 2012;22:1586–93. 15. Adams T, Davidson L, Litwin S, Kolotkin RL, LaMonte MJ, Pendleton RC, et al. Health benefits of gastric bypass surgery after 6 years. JAMA. 2012;11:1122–31. 16. Adams TD, Gress RE, Smith SC, Halverson RC, Simper SC, Rosamond WD, et al. Long-term mortality after gastric bypass surgery. N Engl J Med. 2007;357(8):753–61. 17. Sjöström L, Narbro K, Sjöström CD, Karason K, Larsson B, Wedel H, Swedish Obese Subjects Study, et al. Effects of bariatric surgery on mortality in Swedish obese subjects. N Engl J Med. 2007;357(8):741–52. 18. Suter M, Donadini A, Romy S, Demartines N, Giusti V. Laparoscopic Roux-en-Y gastric bypass: significant long-term weight loss, improvement of obesity-related comorbidities and quality of life. Ann Surg. 2011;254(2):267–73. 19. Buchwald H, Estok R, Fahrbach K, Banel D, Sledge I. Trends in mortality in bariatric surgery: a systematic review and meta-­ analysis. Surgery. 2007;4:621–32. 20. Podnos YD, Jimenez JC, Wilson SE, Stevens CM, Nguyen NT. Complications after laparoscopic gastric bypass: a review of 3464 cases. Arch Surg. 2003;138(9):957–61. 21. King WC, Chen JY, Mitchell JE, Kalarchian MA, Steffen KJ, Engel SG, et al. Prevalence of alcohol use disorders before and after bariatric surgery. JAMA. 2012;307(23):2516–25. Laparoscopic Sleeve Gastrectomy: Technique and Outcomes 13 Natan Zundel, Juan D. Hernandez R., and Michel Gagner Chapter Objectives 1. Describe the surgical technique of laparoscopic sleeve gastrectomy with attention to the steps that prevent complications. 2. Present technical modifications that may represent improvement to the procedure. 3. Summarize the outcomes of laparoscopic sleeve gastrectomy and their relation to surgical technique. Introduction About 18 years ago, laparoscopic sleeve gastrectomy (LSG) was proposed as part of a two-stage procedure in high-risk patients. Today it is not only an accepted stand-alone procedure to effectively treat morbid obesity, but it has become the most frequently practiced bariatric procedure in the USA since 2013 when it overtook gastric bypass with a percentage of 42 versus 34 of the latter [1] and since 2014 worldwide according to an IFSO survey [2]. LSG experts and the bariatric surgical community in several meetings have also endorsed it [3, 4]. These same consensuses have underscored the importance of those procedures being practiced by expert bariatric surgeons. That is true even considering that low morbidity and mortality have been advocated as advantages N. Zundel Department of Surgery, Florida International University Herbert Wertheim College of Medicine, North Miami Beach, FL, USA J. D. Hernandez R. Surgery and Anatomy, Hospital Universitario Fundación Santa Fe de Bogotá, Universidad de los Andes School of Medicine, Bogotá, Colombia M. Gagner (*) Department of Surgery, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA Department of Surgery, Hôpital du Sacré-Coeur, Montreal, QC, Canada over more complex procedures such as laparoscopic Roux-­ en-­Y gastric bypass and biliopancreatic diversion with duodenal switch or other restrictive procedures like the laparoscopic adjustable gastric banding that can have serious anatomical complications in the mid- to long-term follow­up. It also avoids intestinal surgery with associated complications such as internal herniation, small bowel obstruction, micronutrient deficiencies, and malnutrition. Additionally, it is a good alternative when a patient is a transplant candidate or has inflammatory bowel disease, multiple adhesions, previous bowel resection and other conditions that preclude the creation of a Roux-en- Y, or a biliopancreatic diversion. It has also been argued that the follow-up is less demanding than for the aforementioned procedures [4]. Since long-term reports are starting to appear, some of these facts are now supported in the literature. These are some of the reasons that have made LSG a technique that is growing in popularity and is offered by most bariatric centers. The history related to the LSG starts in 1990 with Marceau’s modification of the classic Scopinaro’s biliopancreatic diversion (BPD) by making a parietal cell gastrectomy to reduce the acid load on the ileum in an effort to decrease the incidence of marginal ulcerations [5]. This gastrectomy was vertical and evolved into a loose gastrectomy over a 60-Fr bougie, which was held against the lesser curvature of the stomach far away from the GE junction without mobilization of the antrum. Gagner reproduced this operation laparoscopically in July 1999, hence performing the first laparoscopic sleeve gastrectomy over a 60-Fr bougie with a duodenal switch. Thereafter, LSG alone was initially conceived as the first of two stages of the biliopancreatic diversion with duodenal switch. The rationale was that the procedure would be technically easier and shorter, therefore reducing morbidity and mortality, since the more technically demanding part of the operation would be carried out after a period of weight loss [6, 7]. The idea came from the observation made by the senior author, after recording a higher mortality associated with super-­ super obesity (BMI > 60) in patients undergoing BPD-DS. A two-step surgery approach was used to reduce operating time © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_13 149 150 N. Zundel et al. and surgery complexity [8]. The first step was the LSG, and after several months, the BPD-DS was completed, with a significant reduction in mortality and good results in weight loss [9]. In several patients a steady and surprisingly good weight reduction with the LSG only was noticed, and it was hypothesized that it could become a primary bariatric procedure on its own, a procedure that still left open the possibility of a second intervention in case of unsatisfactory weight loss or weight regain. This suggestion, supported by the author’s own data, was made by other researchers [10]. It became a safe alternative for high-risk patients in need of the operation. LSG is technically easier when compared to gastric bypass or biliopancreatic diversion, a primary reason for its growing popularity among surgeons and patients. It does, however, as with any other surgical procedure, have a potential for long-term complications that range from 0.7% to 6% in different series [11–15]. Some of these complications can be severe and potentially fatal. Therefore, it is important to be thorough in the appropriate technique, to observe the correct steps and safety features, and to follow patients closely after surgery to avoid long-term complications or failure. Preparation and Surgical Technique A multidisciplinary team dedicated to treat obese patients evaluates all patients to ascertain the indication for a bariatric procedure and works toward improving patient diet, physical condition, and habits during the preoperative period. Identified comorbidities are controlled, and patients are fol- Fig. 13.1 (a) Five-trocar setting. (b) Six-trocar setting. The size of the larger trocars depends on the type of stapler to be used a lowed all through the perioperative period. This includes gastroscopy, Helicobacter pylori treatment if present, and respiratory-related measures. Patients are placed in supine, either with legs spread (French position) or closed together (American position). Either way, they are firmly secured to the operating table, to allow for a steep Fowler (reverse Trendelenburg) position. Additionally, the table is slightly tilted right side down for an adequate visualization of the gastroesophageal (GE) junction. Additionally, anti-embolic stockings and intermittent compression devices are employed to prevent venous thromboembolism. Authors’ current LSG technique uses five or six ports. Figure 13.1 shows the options of port distribution. The first trocar, 10–12 mm, is placed at the umbilicus, using an open technique to reach the peritoneal cavity. Two more 5- or 12-mm ports are placed in the supraumbilical region, one subxiphoid and another in the right upper quadrant. Two 15-mm trocars, wide enough to allow the insertion of the largest staplers, are placed in the mid-abdomen just medial to the midclavicular lines. Finally, a 5-mm trocar used by an assistant for retraction is placed in the left upper quadrant, high enough to reach the top of the gastric fundus. A 10-mm, 30° scope is used. The left lobe of the liver is retracted to expose the entire GE junction and the lesser curve. The procedure starts by cutting the small branches of the gastroepiploic arcade and opening the lesser sac. Then, dissection is carried out along the greater curve, staying very close to it, dividing the branches of both gastroepiploic arteries, until short gastric vessels are divided using an advanced bipolar cutting device or the ultrasonic scalpel. The assistant retracts the omentum laterally during the maneuver and b 13 Laparoscopic Sleeve Gastrectomy: Technique and Outcomes keeps repositioning the instrument superiorly to improve exposure of the vessels and avoid bleeding. The remainder of the gastrocolic ligament (without gastroepiploic vessels transection) is severed distally up to 2 cm proximal to the pylorus. The objective of cutting the omentum right by the edge of the greater curve is to minimize the amount of fat attached to the stomach, to make its extraction from the abdomen easier at the end of the operation. The stomach is then lifted to expose its posterior aspect, and all lesser sac attachments to the stomach are freed. This will allow the appropriate positioning of the mechanical suture and avoid bleeding. When cutting these adhesions, it is necessary to be aware of the presence of the branches of the left gastric artery, identifying and preserving them. If the left gastric branches were cut, the blood supply to the sleeve would be compromised. Other anatomic relations the surgeon needs to be aware of are the splenic artery and vein, running along the superior edge of the pancreas. Splenic artery in older patients may be redundant and therefore may be in harm’s way during posterior dissection and stapling. The gastrophrenic ligament is divided, and the fat pad usually found anterior to the cardia is removed to improve exposure of the angle of His, adding the full exposure of the left crus to complete the dissection and to determine the presence of a hiatal hernia. In case such a hernia is discovered, a formal closure of the hiatus is mandatory, since gastroesophageal reflux disease (GERD) and other complications are associated with its presence in the postoperative period. To achieve that, the stomach involved and distal esophagus are freed of mediastinal attachments and brought down into the abdomen, leaving at least 3 cm of intraabdominal esophagus. A posterior crural approximation is conducted to close the gap, using nonabsorbable sutures and pledgets if needed (Fig. 13.2). It has been proposed in the literature that GERD with or without a hiatal hernia can be treated or prevented. To Fig. 13.2 Laparoscopic posterior closure of a hiatal defect is required in patients with hiatal hernia or GERD 151 that purpose and as mentioned ahead, a modified Hill repair has been advocated by some surgeons to keep the sleeve in the abdominal cavity. Stomach division starts 4 cm proximal to the pylorus, to preserve a part of the gastric emptying mechanism of the antrum. Prior to the creation of the sleeve, the anesthetist introduces a 34–40-Fr bougie to guide the stapling and maintain an adequate lumen of the gastric sleeve. Continuous communication between surgeon and anesthetist is paramount to ensure adequate positioning of the bougie in a safe fashion. The bougie should be placed prior to stapling, guiding it to reach the pylorus, and positioned close to the lesser curve. Care is taken not to divide the stomach too close to the incisura angularis to avoid kinking or stenosis at this level. Green (4.8 mm) or black (5 mm) stapler cartridges are used with absorbable buttress material (Gagner). Green or black for the first two firings and blue or gold for the rest if no absorbable buttressing materials are used (Zundel). In any case, all of them are 60 mm in length. Stapling is performed in a way that no kinking or twisting of the sleeve is produced at any level. To achieve this, the stomach is held by the assistant stretching it to the patient’s left, while the surgeon places the stapler making sure that anterior and posterior edges are at the same distance from the lesser curve. In other words, the distance of the anterior aspect of the remaining stomach should not be shorter than its posterior counterpart. Additionally, a stapler should be placed right at the angle of the previous one, avoiding “dog-ears” created on the edge of the stomach that may produce ischemia. After each firing, the anesthetist is asked to wiggle the bougie to ensure the sleeve is not too tight or that the bougie has not been stapled or cut. Although the senior author recommended in the past to cut the fundus at least 1 cm from the gastroesophageal junction, his current practice is to divide it as close as the GE junction as possible, without actually compromising the esophagus. The other authors still cut the fundus 0.5 cm away from the GE junction and imbricate the staple line with absorbable suture in an effort to reduce leak rates. This is done without the presence of buttressing material. The senior author’s intention is to create a sleeve that goes in straight line from the GE junction down into the stomach, since a funnel-shaped sleeve may be more likely to produce gastroesophageal reflux by dilatation and stretching of the lower esophageal sphincter. Additionally, the perigastric fat is mobilized, permitting better identification of the esophagogastric junction, and this may be used to buttress the staple line. In this technique, the staple line is reinforced only at the GE junction, where leaks are more frequent, and at the bottom of the staple line on the antrum, the thickest part of the stomach. This is done using through-and-through figure-­ eight stitches with 3–0 absorbable monofilament sutures. 152 N. Zundel et al. Fig. 13.3 Reinforcement of the stapler line with running suture. Only if the stapler line does not have buttressing materials The other authors (Zundel and Hernandez) do not routinely use absorbable buttressing material; conversely, with the bougie in place, the full length of the staple line is oversewn with a running suture of 3–0 absorbable suture (Fig. 13.3). The anesthetist removes the bougie under direct vision to check the final shape of the sleeve. The stomach is removed through one of the 12–15 mm ports (Fig. 13.4). The integrity of the staple line is tested with the instillation of 50–100 ml of methylene blue in saline solution. No drains are left. Postoperative Period Appropriate hydration and pain and nausea control is initiated. During in-hospital stay, patients are observed for signs of leak or bleeding such as tachycardia, tachypnea, or fever. Abdominal pain and left shoulder pain are not reliable symptoms at this point, but should not be dismissed as normal. Anti-embolic stockings and intermittent sequential compression devices can be removed as soon as the patient is ready to walk. The next day, an upper gastrointestinal contrast X-ray is done to identify any possible leaks (Fig. 13.5). If the study is negative for leaks, liquid diet is started, and patients are encouraged to ambulate. Respiratory therapy is initiated and previous chronic medication is restarted. If early signs suggesting a leak appear, the patient is taken to surgery for a diagnostic laparoscopy. Patients are usually discharged home on the first or second postoperative day with liquid pain medications for a few days and a proton pump inhibitor for 6–8 weeks. Also, patients are initiated in a liquid diet for 2 weeks, followed by a semiliquid diet following the first, until progressively more solid diet is allowed. A team of dietitian, surgeon, nurse, and according to need a psychiatrist, internist, or other specialist follows patients. Results Weight Loss and Comorbidities A number of reports on outcomes of LSG with patients followed for more than 5–10 years are being published more often, giving the scientific community more data to make possible to reach conclusions about the safety, efficacy, and durability of LSG. However, it is important to point out that the large number of variations that still exist in surgical technique may be cause for confusion and difficulty in establishing comparable outcomes at the present time [15]. Most patients with longer follow-up were operated at a time when surgical technique was even less standardized. The bariatric community has made an effort to come to an agreement in major technical issues through consensus meetings on LSG. Five of these meetings explored the opinions of experts and the evidence in the literature, creating concurrence that has reduced technical variations in topics such as bougie size, starting point of stapling, etc. [4]. Recommendations have been made, and a more homogeneous technique has been developed. These consensus meetings started in 2007, so as mentioned before, outcomes that could be attributable to these agreements are only coming to be available now. 13 Laparoscopic Sleeve Gastrectomy: Technique and Outcomes 153 Fig. 13.4 Shape of the resected stomach with a continuous staple line and an additional resection of the fundus. On the right the stomach is insufflated to see its size and shape. Left: regular stapler line. Right: staple line reinforced with buttressing material Fig. 13.5 Upper GI contrast study showing uniform shape of the sleeve with no evidence of leaks and good flow through the sleeve The LSG summit of 2012 [16] reported on a survey answered during the meeting by 130 surgeons with experience of more than 1 year doing the operation, with a total of 46,133 LSGs. The survey included surgeons with short experience and minimum follow-up. A calculation on what surgeons reported rendered a mean % excess weight loss (EWL) of 59.3% in year 1, 59.0% in year 2, 54.7% in year 3, 52.3% in years 4 and 5, and 50.6% in year 6 [16]. The authors recommend caution when analyzing these numbers, since they determined that surgeons marked 0 change in EWL% when they should have left a blank for not having patients that far in time. Since it was not possible to discard the 0% EWL option, they did not eliminate those numbers, but adjusting the analysis for this bias, % EWL could be even higher. On 2014, the Experts Survey carried out before the fifth consensus in Montreal reported at 5-year follow-up an EWL of 60.5%, with an average patient BMI post-surgery of 30.2 kg/m2. The average rate of strictures was 2.1% and the leak rate reported was 2.4%. In what concerns to conversions, 4.7% were due to weight loss failure and 2.9% due to GERD [4]. 154 Studies with a long-term follow-up support better results in weight loss than those reported by the survey. Bohdjalian et al. found a 5-year % EWL of 54.8 ± 6.9, which was comparable to the results at 1 year, commenting that LSG leads to stable weight loss in the long-term follow-up [17]. In a study with a large number of super obese patients that extended follow-up to 3 and 5 years, Saif et al. showed that the percentage of excess BMI lost was maintained. The mean percentage of excess BMI lost was statistically significant for all cohorts, being 58.5% at 1 year, 65.7% at 3 years, and 48% at 5 years [18]. Zachariah et al. report the data collected from 228 patients treated with LSG and followed for 5 years since 2007. They showed a mean % EWL of 71.2 ± 21 at 3 years and 63 ± 20 at 5 years, with BMI going down to 26 and 28, respectively. Mortality was reported at 0.43% [19]. It is worth pointing out as a parenthesis that in high-volume centers mortality is even lower [13, 14]. At 5 years, resolution of diabetes was 66%, 50% for hypertension, and 100% for hyperlipidemia. In fact, results for diabetes resolution have been found to be as good as that of laparoscopic Roux-­ en-­Y gastric bypass [20]. Two recent studies have published more than a thousand patients, each with more than 5 years follow-up. Alvarenga et al. [13] reported 1020, some of them operated up to 8 years before. EWL for this series was 92% after 1 year, 89% after 3 years, 75% after 5 years, and 73% after 8 years. The caveat was a large desertion of patients as the follow-up period was longer, up to 92% at 8 years. Authors do not give the actual number, making difficult to judge these results; however, they argue that the main power of the paper is in the 3–5 years follow-up and the large number of patients in their cohort. In the second paper, van Rutte et al. [14] reported on 1041 patients followed for up to 5 years. Percentage of EWL was 69.3% at 3 years, 70.5 at 4 years, and 58.3 at 5 years. Both studies consider LSG a safe and effective treatment for morbid obesity. Complications are low and comorbidities control is very good. They also showed that mean EWL after 5 years remains over 50%, with the overall percentage in 70%. Interestingly, van Rutte also found that of the 16.4% of patients who reported GERD before surgery, complaints disappeared in 84.5%. On the other hand, 11.7% developed de novo reflux disease. In relation with the metabolic effects of LSG, there was a concept that therapeutic results were achieved only by reducing the stomach size approximately to 80% of its original size. Therefore, the resulting weight loss was a consequence of a mechanical or volumetric variation. This concept has been abandoned as a number of studies have shown other possible causes. There are indeed humoral changes that impact metabolism, therefore affecting weight. Several studies have shown that after the operation, plasma ghrelin levels were significantly reduced in the early postoperative period [21, 22] and remain consistently low during 5-year follow-up studies N. Zundel et al. [17]. This peptide, produced mainly in the stomach, is secreted when the stomach is empty, increasing hunger and producing secretions to digest food. Another molecular effect that is creating interest among researchers is the interaction between LSG, bile acid circulation, and FXR signaling [23]. It has been reported that levels of circulating bile acids are higher after LSG [24]. This has an impact on gut microbiota, contributing to the changes that have been associated with weight loss. The Farsenoid X receptor, also known as NR1H4, is a nuclear receptor and, when it binds to bile acids, creates signaling molecules that take part in regulating several metabolic processes. It has been suggested that these metabolic processes have great impact in weight reduction, possibly more than the stomach volume restriction itself [24]. A general belief of minimal nutritional deficiencies has accompanied the practice of LSG, which originated in the fact that nutrients follow the normal path in the gastrointestinal tract, as opposed to gastric bypass and biliopancreatic diversion, where large segments of the small intestine are excluded from the passage of nutrients. There is still insufficient documentation available to support or dismiss this assumption. Gehrer et al. [25] found nutritional deficiencies both before gastric bypass and after LSG with a mean follow-­up of 24 months. Deficiencies in zinc, vitamin D3, folic acid, iron, and vitamin B12 were present but not clinically significant and less important than after gastric bypass, and all were easily treated and resolved. A 5-year follow-up study showed a different picture, with values for parathyroid hormone, hemoglobin, and hematocrit just under the normal values but with no deficiencies [18]. For the authors, the results show clear health improvements with nutrient indicator levels reaching up to normal values with no signs of nutritional deficiencies and conclude that long-term follow-up is fundamental to assist patients in maintaining the good weight loss results. Complications On the large series by Alvarenga [13], 39 patients (3.8%) had long-term morbidity that included strictures in 5 of them (0.49%) and GERD in 61 (6%). The overall 30-day mortality rate was 0%. Conversion rate to LRYGB was 1.25% (n = 12), five of them (0.49%) due to intractable GERD, four (0.43%) for weight regain, and three (0.29%) for gastric outlet obstruction. In the group of van Rutte et al. [14], complications rates decreased with improved technique over time. No death was attributed directly to surgery. Of 724 patients, 11.7% developed GERD de novo, and 8.2% presented over time with dysphagia leading to vomiting. One of the most feared complications, fortunately rare, is leaks. However, fistula, stenosis, GERD, and pouch dilata- 13 Laparoscopic Sleeve Gastrectomy: Technique and Outcomes tion, among others, also can be present [11]. Leakage usually appears as an acute complication (within 7 days), causing tachycardia, tachypnea, and fever very early on, and indicates most often that the patient requires immediate intervention. The most common site for leak is along the staple line immediately below the gastroesophageal junction. Several strategies can be used including diagnostic laparoscopy with drainage, insertion of a T-tube in the opening to control the fistula, insertion of an esophagogastric stent to occlude the perforation and to open any associated distal narrowing, or percutaneous drainage with endoscopic stents. Some European groups have used endoluminal double pigtail catheters to the same end. The most frequent location of the fistula is near the angle of His (at the top of the stapled line) and secondly in the antrum at the beginning of the gastric stapled line. Sometimes fistulae can become chronic and therefore will need a different and more complex treatment [26]. A laparoscopic Roux-en-Y fistula-jejunal anastomosis can be performed as early as several weeks after a leak, with a high success rate, thus avoiding total gastrectomy, a procedure with higher morbidity. Another complication and common cause for leakage is stricture or stenosis at the level of the gastric incisura [27], a cause of obstruction. Clinical presentation both for acute and late LSG obstruction is similar with dysphagia appearing weeks to months after the LSG operation in the latter, starting with dysphagia to solids followed by symptoms to liquids, salivation, and vomiting. Endoscopy with balloon dilatation is the preferred method for management of the stricture. Zundel et al. recommend an achalasia balloon with higher controlled pressure [26]. Acute obstruction cases may be due to gastric mucosal edema and external compression and in some cases due to kinking of the sleeve [9]. Cottam et al. noted that kinking is independent of bougie size and more correlated with the oversewing of the suture line rather than with the diameter of the sleeve [27]. Stricture can be avoided by keeping a safe distance from the bougie at the level of the incisura angularis. This also applies when the staple line is oversewn to reinforce it. All along the staple line, the bougie will prevent stitching through the stomach’s lumen or further reducing the size of the sleeve. In all these cases, kinking may appear. The recommended bougie size is 36 F [28], as nearly 40% of surgeons have used this size [16]. The alteration of the pouch architecture caused by asymmetric stapling of the anterior and posterior walls of the stomach may lead to twisting of the gastric tube, which may also cause dysphagia [29]. Also, leaving an excessively large posterior wall may twist the sleeve as the stapler is applied, leaving an uneven line of staples. If a stricture becomes permanent and does not respond to endoscopic treatment such as dilatation or stent, then a laparoscopic longitudinal lateral gastrotomy with transverse hand-sewn closure (like a Mikulicz pyloroplasty) can be performed. Alternatively an anterior 155 seromyotomy, rarely performed due to the risk of mucosal perforation and leak, or conversion to Roux-en-Y gastric bypass can be performed. The association between GERD and LSG has been one of the most controversial issues of the technique. There are multiple causes for pathological reflux in obese patients, even before surgery. Increased intraabdominal pressure and a higher rate of hiatal hernia are two of them [30]. De novo GERD can appear, arguably more after LSG, but also after RYGBP, and if a hiatal hernia is left untreated at the time of the operation, the chances of severe or even intractable reflux are important. Several studies have shown an incidence between 6% and 16% [13]. On the other hand, Lyon et al. showed an improvement of GERD if hiatal hernia was repaired with anterior and posterior hiatal closure [31] in patients with history of GERD and hiatal hernia diagnosed both pre- or intraoperatively. They also reported a slight but not statistically significant rise in reflux symptoms in patients previously asymptomatic and without hiatal hernia. These results must be analyzed with caution since it is a retrospective study and patients were only interviewed by phone. The discussion started when cases of GERD appeared in relation to LSG [27], blaming the operation for all of them. It was then reported up to 79% of heartburn in obese patients in the preoperative period, with regurgitation in 63%. Additionally near half of them had objective GERD as demonstrated by esophagitis at endoscopy in almost a fifth of the total [32]. Although several authors attribute mechanisms for de novo reflux to the destruction of the phrenoesophageal membrane, the section of the sling fibers at the cardial region and the disappearance of the angle of His [32], others consider that it is in the technique of the creation of the gastric tube where causes can be found [33]. A review of the literature in 2015 showed great variability in results, with some papers showing high incidence of postoperative reflux either worsening or appearing de novo, while other talked of reduction or absence of the GERD symptoms present in the preoperative period [34]. It was true by the time of that publication as it is now that there are no valid studies that shed light on this topic. On a review of his own experience on a period of 18 months, and after 6 years of practicing LSG, Daes et al. give a different picture [33]. They prospectively followed 373 of 382 consecutive patients undergoing LSG for up to 22 months, at least 97% of them more than 6 months. Using endoscopy, they found GERD in 44.5% of patients and hiatal hernia in 51%. The hernia was confirmed during surgery in 37.2% of patients. According to them, with a very well-­standardized technique, plus being aggressive in the search and surgical correction of hiatal hernia, they achieved a very low incidence of postoperative GERD. They did not use pH analysis or manometry; however the incidence of GERD on 373 patients was only 2.6%, of the patients with preoperative GERD symptoms 94% 156 were asymptomatic, and only one patient of the previously asymptomatic patients reported GERD complaints. A very recent meta-analysis [35] produced a recidivism rate between 14% and 37% and a revision rate close to 20%, mostly due to weight regain, and in a small percentage of near 3%, to GERD. Authors discussed that findings are from 7 to 11 years ago, and therefore surgeon’s experience, standardized technique, and thorough follow-up were not there yet, and therefore there is still a lot of variation in results. That is also true, they say, when revisions are analyzed. Variations in revision rates and their causes are very high, suggesting differences in experience and technique. Most of patients with early postoperative symptoms of GERD respond to prolonged medical therapy; however, some patients require surgical reoperation with hiatal hernia repair, or conversion to Roux-en-Y gastric bypass, which has been advocated as the best alternative in these cases. Hiatal hernia repair with the use of nonabsorbable mesh is avoided due to the risk of erosion into the gastric lumen. Current recommendations are to reduce the hernia with an appropriate length of intraabdominal esophagus and closure of the hiatus, which most surgeons tailor posteriorly [36]. Recent publications advocate for what some authors have called a modified Hill technique, in reference to Lucius Hill anterior fundoplication with a gastropexy to the arcuate ligament [37]. A French group has used the technique since December 2014 [38]. The first 14 patients they described, all symptomatic, had a mean excess BMI loss of 68%. There were no major complications. Of three patients complaining of persistent GERD symptoms, only one required daily use of PPIs. They have called their technique simplified laparoscopic Hill repair. The authors consider posterior gastropexy a more appropriate name for it, since no fundoplication is possible. It has been used both at the first operation and as a procedure to correct GERD and hernia [39]. In spite of all measures possible, there will be failures, and LSG should in this case be considered as a first step treatment in obese patients and be converted into a second procedure [15]. Weight regain, GERD, inadequate weight loss, or inadequate resolution of comorbidities may require late reintervention, after proper endoscopic and radiological evaluation. Choice of operation will be dependent on the initial BMI (super obese will respond better with a duodenal switch), presence or absence of GERD (Roux-en-Y gastric bypass is preferred), or re-sleeve gastrectomy for fundus dilatation. Sleeve Gastrectomy or Gastric Bypass? Since it is the gold standard of bariatric surgery, it is inevitable to compare RYGBP with today’s more frequently practiced operation for obesity: sleeve gastrectomy. Gastric N. Zundel et al. bypass has been practiced for more than 50 years; although several modifications have been done along the years and different groups have introduced variations, the technique has been fairly standardized for several decades. The first series of LRYGBP was published in 1994, and since then, minimally invasive approach became the preferred route. On the other hand, LSG’s longer meaningful series are reaching only 8 or 9 years, and therefore comparisons that make justice to both procedures are still scarce. SM-BOSS [40] is a Swiss multicenter randomized trial that compares LSG and LRYGBP. Both procedures are standardized in the four hospitals that are collecting data. They recently published an update with 5-year follow-up that shows no difference in weight loss between both procedures. There is a slight increase in excess BMI every year for both procedures; however, the results remain successful for both: 61.1% excess BMI loss for LSG and 68.3% for LRYGBP, both good and not significantly different (P = 0.22). On secondary observations, complete remission of diabetes was seen in 61.5% and 67.9%, respectively (P = 0.99); total remission of dyslipidemia was significantly higher in LRYGBP than in LSG (62 vs. 42%, P = 0.09). In reference to a complication extensively exposed before, they found remission of preexisting GERD after 5 years in 25% of patients after LSG and 60% after LRYGBP (P = 0.002). They also reported higher worsening of symptoms after LSG (31.8% vs 6.3% P = 0.006); additionally, de novo GERD was found in 31.6% after LSG and 10.7% after LRYGB (P = 0.01). However, other complications were significantly less frequent in LSG. Both techniques are nowadays and in experienced groups very safe, but there were more reinterventions in the 30-day postoperative period, as there were late complications like internal hernia and obstruction and dumping, among others. Additionally, there was one dead on the LRYGBP group only [40]. SLEEVEPASS [41], also a multicenter randomized clinical trial, this one conducted in Finland, has now 5-year follow-­up results published. It was designed to establish if there was equivalence between LRYGBP and LSG. Two hundred forty patients were involved. In it, percentage of EWL at 5 years was higher for LRYGBP, but the difference was not statistically significant. As authors report, “gastric bypass resulted in statistically greater weight loss than sleeve gastrectomy at 5 years, but the difference was not clinically significant, as the minimal clinically important difference of 9 is within the confidence interval” [41]. Diabetes remission rate was lower than in the SM-BOSS trial, and no difference was found between both procedures. A remarkable finding in complications was that de novo GERD was only 9.1% after 30 days postoperative of LSG and only 5.8% at long-term follow-up. In terms of reintervention, GERD was associated with 7 of 10 surgeries. No mention was made of the relation between GERD 13 Laparoscopic Sleeve Gastrectomy: Technique and Outcomes 157 and RYGBP. Additionally, there was no difference in patients, it is going to be the only intervention they need, as quality of life between both groups. long as they have discipline and a good team to support them The merit of SLEEVEPASS and SM-BOSS is high-­ in the process, among other things. quality evidence, but other papers have shown comparable However, it may have come the time to consider obesity results in Israel, Korea, and the USA [42–44]. In a large as other chronic diseases are approached, like hypertension population study that included surgical and nonsurgical or diabetes, where treatment has to be planned in stages that patients, after a mean 4.5 year follow-up, Reges et al. [42] grow in complexity. Consequently, for obesity, surgery and reported a higher survival rate in surgical as compared to endoscopic interventions will have to be part of those steps, nonsurgical patients. In the surgical group, comprising 8385 and possibly patients will have to be informed that a second patients, mortality was significantly lower among LSG operation may be needed in the future. In such scenario, LSG patients, compared to LRYGBP and adjustable gastric band- is probably the most versatile operation: it will be more than ing. Revisional surgery was a less frequent in the first two enough for most patients but can also be the best first interprocedures and higher in gastric banding. In a study with vention in a stepped program, since it can be redone or conelderly patients comparing both techniques, Janik et al. [43] verted to RYGBP, BPD-DS, or SADIs. reviewed the Metabolic and Bariatric Surgery Accreditation and Quality Improvement Program (MBSAQIP) database of 2015 and selected 6742 patients over 60 years that ­underwent Conclusions one of the two bariatric procedures and matched them 1 to 1. Compared to LSG, LRYGB had significantly higher anasto- The advantages of LSG are still being confirmed today, but motic leakage rate (1.01% vs. 0.47%, P = 0.019), more fre- also placed in perspective in the whole spectrum of obesity quent 30-day readmission rate (6.08% vs. 3.74%, P < 0.001), surgery. This procedure has earned an important place in barand higher 30-day reoperation rate (6.08% vs. 3.74%, iatric surgeons’ armamentarium and has become the most P < 0.001). There was no significant difference in bleeding, practiced intervention as a stand-alone operation. infection, sepsis, and pulmonary embolism, among others. If one considers that LSG’s surgical technique has only Kim found no significant difference in the mean percentage been well defined in the last 5 or 7 years, it is possible to of EWL between LSG (63.5%) and LRYGBP (71.2% hypothesize that outcomes reported in the future may be P = 0.073), although revision was higher in LSG [44]. even better than those currently found in literature. This, of course, will require good patient selection, expert multidisciplinary teams, and adequate long-term follow-up. Discussion One fundamental consideration that has reached consensus among experts is that GERD and hiatal hernia must be Severe obesity should be considered a chronic and mostly carefully sought after, both in the preoperative and postopincurable disease influenced by many features and with erative periods, and if found, treated aggressively. There are many etiological factors. In fact, emotional and psychologi- suggestions that an appropriate surgical technique may cal components in obesity are paramount, not only as a cause reduce its incidence. for obesity itself but also as a reason of failure of surgical and Finally, there is still work to be done in standardizing sevnonsurgical treatments. It is worth considering that a multi- eral important steps in the surgical technique, as there is still factorial chronic disease, with a considerable behavioral controversy in bougie size, suture reinforcement, and how component, is not best treated with surgery. However, after much of the antrum is to be resected. several decades, surgery is still the best method to achieve such a large rate of success, that is, a significant and durable weight loss and control of comorbidities. However, an Question Section important and growing number of patients have some sort of failure. And that is true for all procedures and surgical 1. What was the origin of the sleeve gastrectomy as such? A. Doctor Gagner’s first staged duodenal switch starting techniques. with a laparoscopic sleeve gastrectomy. One of the items growing more in bariatric surgery today B. Doctor Marceau’s modification of Scopinaro’s classic is reinterventions. A larger number of patients are going for biliopancreatic diversion creating a vertical a second operation every year, and this can only be expected gastrectomy. to grow, as the rate of operations is higher. LSG has demonC. A consensus of bariatric surgery experts created a verstrated to be a safe, reproducible, and effective operation on tical gastrectomy by removing fundus and body of the its own to treat morbid obesity with a rate of success that is, stomach. arguably, at least as good as the Roux-en-Y gastric bypass, D. Mason did the first sleeve gastrectomy. according to several studies. For a large percentage of 158 2. What are the main measures if a hiatal hernia is found? A. The hiatus is not explored; only the fundus is freed and prepared for resection. B. The procedure is abandoned and the patient informed of the options. C. Reduction of the hernia with 3 cm of intraabdominal esophagus and crural approximation. D. Create a formal fundoplication to prevent GERD. 3. Metabolic effects of LSG are mainly related to the reduction of the production of: A. Insulin B. GLP-1 C. Ghrelin D. Glucose 4. Where are leaks after LSG more frequently located? A. Staple line below gastroesophageal junction B. Staple line at the start near the pylorus C. Staple line at the incisura angularis D. Caused by accidental perforation anywhere in the stomach References 1. Ponce J, DeMaria EJ, Nguyen NT, Hutter M, Sudan R, Morton JM. American Society for Metabolic and Bariatric Surgery estimation of bariatric surgery procedures in 2015 and surgeon workforce in the United States. Surg Obes Relat Dis. 2016;12:1637–9. 2. Angrisani L, Santonicola A, Iovino P, Vitiello A, Zundel N, Buchwald H, Scopinaro N. Bariatric surgery and endoluminal procedures: IFSO worldwide survey 2014. Obes Surg. 2017;27:2279–89. 3. Rosenthal R. International sleeve gastrectomy expert panel consensus statement: best practice guidelines based on experience of 12,000 cases. Surg Obes Relat Dis. 2012;8:8–19. 4. Gagner M, Hutchinson C, Rosenthal R. Fifth international consensus conference: current status of sleeve gastrectomy. Surg Obes Relat Dis. 2016;12:750–6. 5. Marceau P, Biron S, Bourque RA, Potvin M, Hould FS, Simard S. Biliopancreatic diversion with a new type of gastrectomy. Obes Surg. 1993;3(1):29–35. 6. Ren C, Patterson E, Gagner M. Early results of laparoscopic biliopancreatic diversion with duodenal switch: a case series of 40 consecutive patients. Obes Surg. 2000;10:514–23. 7. Regan J, Inabnet W, Gagner M. Early experience with two-stage laparoscopic Roux-en-Y gastric bypass as an alternative in the super-super obese patient. Obes Surg. 2003;13:861–4. 8. Kim WW, Gagner M, Kini S, et al. Laparoscopic versus open biliopancreatic diversion with duodenal switch: a comparative study. J Gastrointest Surg. 2003;7:552–7. 9. Gagner M, Inabnet WB, Pomp A. Laparoscopic sleeve gastrectomy with second stage laparoscopic biliopancreatic diversion and duodenal switch in the superobese. In: Inabnet WB, EJ DM, Ikramuddin S, editors. Laparoscopic bariatric surgery. Philadelphia: Lippincott, Williams & Wilkins; 2005. 10. Mognol P, Chosidow D, Marmuse JP. Laparoscopic sleeve gastrectomy as an initial bariatric operation for high-risk patients: initial results in ten patients. Obes Surg. 2005;15:1030–3. 11. Frezza EE, Reddy S, Gee LL, Wachtel MS. Complications after sleeve gastrectomy for morbid obesity. Obes Surg. 2009;19:684–7. N. Zundel et al. 12. Lalor PF, Tucker ON, Szomstein S, Rosenthal RJ. Complications after laparoscopic sleeve gastrectomy. Surg Obes Relat Dis. 2008;4:33–8. 13. Alvarenga ES, Lo Menzo E, Szomstein S, Rosenthal RJ. Safety and efficacy of 1020 consecutive laparoscopic sleeve gastrectomies performed as a primary treatment modality for morbid obesity. A single-center experience from the metabolic and bariatric surgical accreditation quality and improvement program. Surg Endosc. 2016;30:2673–8. 14. van Rutte PWJ, Smulders JF, de Zoete JP, Nienhuijs SW. Outcome of sleeve gastrectomy as a primary bariatric procedure. British J Surg. 2014;101:661–8. 15. Noel P, Nedelcu M, Eddbali I, Manos T, Gagner M. What are the long-term results 8 years after sleeve gastrectomy? Surg Obes Relat Dis. 2017;13(7):1110–5. 16. Gagner M, Deitel M, Erickson AL, Crosby RD. Survey on laparoscopic sleeve gastrectomy (LSG) at the fourth international consensus summit on sleeve gastrectomy. Obes Surg. https://doi. org/10.1007/s11695-013-1040-x. Published on-line on 04/08/2013. 17. Bohdjalian A, Langer FB, Shakeri-Leidenmuhler S, Gfrerer L, Ludvik B, Zacherl J, Prager G. Sleeve gastrectomy as sole and definitive bariatric procedure: 5-year results for weight loss and ghrelin. Obes Surg. 2010;20:535–40. 18. Saif T, Strain GW, Dakin G, Gagner M, Costa R, Pomp A. Evaluation of nutrient status after laparoscopic sleeve gastrectomy 1, 3 and 5 years alter surgery. Surg Obes Relat Dis. 2012;8:542–7. 19. Zachariah SK, Chang PC, Ooi AS, et al. Laparoscopic sleeve gastrectomy for morbid obesity: 5 years experience from an Asian center of excellence. Obes Surg. 2013;23:939–46. https:// doi.org/10.1007/s11695-013-0887-1. 20. Yip S, Plank LD, Murphy R. Gastric bypass and sleeve gastrectomy for type 2 diabetes: a systematic review and meta-analysis of outcomes. Obes Surg. https://doi.org/10.1007/s11695-013-1030-z. Published on-line 17/08/2013. 21. Sanchez-Santos R, Masdevall C, Baltasar A, et al. Short- and mid- term outcomes of sleeve gastrectomy for morbid obesity: the experience of the Spanish National Registry. Obes Surg. 2009;19:1203–10. 22. Anderson B, Switzer NJ, Almamar A, Shi X, Birch DR, Karmali S. The impact of laparoscopic sleeve gastrectomy on plasma ghrelin levels: a systematic review. Obes Surg. 2013;23:1476–80. https://doi.org/10.1007/s11695-013-0999-7. 23. Ryan KK, Tremaroli V, Clemmensen C, et al. FXR is a molecular target for the effects of vertical sleeve gastrectomy. Nature. 2014. 8;509:183–8. 24. Myronovych A, et al. Vertical sleeve gastrectomy reduces hepatic steatosis while increasing serum bile acids in a weight-loss-­ independent manner. Obesity. 2014;22:390–400. 25. Gehrer S, Kern B, Peters T, Christofel-Courtin C, Peterli R. Fewer nutrient deficiencies after laparoscopic sleeve gastrectomy (LSG) than after laparoscopic Roux-Y-gastric bypass (LRYGB)-a prospective study. Obes Surg. 2010;20:447–53. 26. Zundel N, Hernandez J. Revisional surgery after restrictive procedures for morbid obesity. Sur Laparosc Endosc Percutan Tech. 2010;20:338–43. 27. Cottam D, Qureshi F, Mattar S, et al. Laparoscopic sleeve gastrectomy as an initial weight-loss procedure for high-risk patients with morbid obesity. Surg Endosc. 2006;20:859–63. 28. Gagner M. Leaks after sleeve gastrectomy are associated with smaller bougies: prevention and treatment strategies. Surg Laparosc Endosc Percutan Tech. 2010;20:166–9. 29. Kakoulidis TP, Karringer A, Gloaguen T, Arvidsson D. Initial results with sleeve gastrectomy for patients with class I obesity (BMI 30–35 kg/m2). SOARD. 2009;5:425–8. 13 Laparoscopic Sleeve Gastrectomy: Technique and Outcomes 30. Pandolfino JE, El-Serag HB, Zhang Q, Shah N, Ghosh SK, Kahrilas PJ. Obesity: a challenge to esophagogastric junction integrity. Gastroenterology. 2006;130:639–49. 31. Lyon A, Gibson SC, De-loyde K, Martin D. Gastroesophageal reflux in laparoscopic sleeve gastrectomy; hiatal findings and their management influence outcome. Surg Obes Relat Dis. 2015;11: 530–7. 32. Braghetto I, Lanzarini E, Korn O, et al. Manometric changes of the lower esophageal sphincter after sleeve gastrectomy in obese patients. Obes Surg. 2010;20(3):357–62. 33. Daes J, Jimenez ME, Said N, Dennis R. Laparoscopic sleeve gastrectomy: symptoms of gastroesophageal reflux can be reduced by changes in surgical technique. Obes Surg. 2012;22:1874–9. 34. Melissas J, Braghetto I, Molina JC, Silecchia G, Lossa A, Iannelli A, Foletto M. Gastroesophageal reflux disease and sleeve gastrectomy. Obes Surg. 2015;25:2430–5. 35. Clapp B, Wynn M, Foster C, O’Dell M, Tyroch A. Long term (7 or more years) outcomes of the sleeve gastrectomy: a meta – analysis. Surg Obes Relat Dis. 2018;14:741–7. 36. Mahawar KK, Carr WRJ, Jennings N, Balupuri S, Small PK. Simultaneous sleeve gastrectomy and hiatus hernia repair: a systematic review. Obes Surg. 2015;25:159–66. 37. Sánchez-Pernaute A, Talavera P, Pérez-Aguirre E, Domínguez-­ Serrano I, Rubio MA, Torres A. Technique of Hill’s gastropexy combined with sleeve gastrectomy for patients with morbid obesity and gastroesophageal reflux disease or hiatal hernia. Obes Surg. 2016;26:910–2. 38. Gero D, Ribeiro-Parenti L, Arapis K, Marmuse JP. Sleeve gastrectomy combined with the simplified Hill repair in the treatment of 159 morbid obesity and gastro-esophageal reflux disease: preliminary results in 14 patients. World J Surg. 2017;41:1035–9. 39. Nassar R, Linares JD, Girón F, Torres E, Zundel N, Hernandez JD. Hiatal hernia correction: a different approach for patients with previous sleeve gastrectomy. Surg Endosc. 2017;31(Suppl 1):S234. 40. Peterli R, Wölnerhanssen BK, Peters T, Vetter D, Kröll D, Borbély Y, Schultes B, Beglinger C, Drewe J, Schiesser M, Nett P, Bueter M. Effect of laparoscopic sleeve gastrectomy vs laparoscopic Roux-en-Y gastric bypass on weight loss in patients with morbid obesity the SM-BOSS randomized clinical trial. JAMA. 2018;319(3):255–65. 41. Salminen P, Helmiö M, Ovaska J, Juuti A, Leivonen M, Peromaa-­ Haavisto P, Hurme S, Soinio M, Nuutila P, Victorzon M. Effect of laparoscopic sleeve gastrectomy vs laparoscopic Roux-en-Y gastric bypass on weight loss at 5 years among patients with morbid obesity the SLEEVEPASS randomized clinical trial. JAMA. 2018;319:241–54. 42. Reges O, Greenland P, Dicker D, Leibowitz M, Hoshen M, Gofer I, Rasmussen-Torvik LJ, Balicer RD. Association of bariatric surgery using laparoscopic banding, Roux-en-Y gastric bypass, or laparoscopic sleeve gastrectomy vs usual care obesity management with all-cause mortality. JAMA. 2018;319(3):279–90. 43. Janick M, Mustafa RR, Rogula TG, Saleh AA, Abbas M, Khaitan L. Safety of laparoscopic sleeve gastrectomy and Roux-en-Y gastric bypass in elderly patients – analysis of the MBSAQIP. Surg Obes Relat Dis. 2018; https://doi.org/10.1016/j.soard.2018.04.008. 44. Park JY, Kim YJ. Laparoscopic gastric bypass vs sleeve gastrectomy in obese Korean patients. World J Gastroenterol. 2015;21(44):12612–9. Biliopancreatic Diversion with Duodenal Switch: Technique and Outcomes 14 Ranjan Sudan Chapter Objectives 1. Understand the history of evolution of the duodenal switch operation. 2. Become familiar with the principles of performing the duodenal switch operation. 3. Understand comparative effectiveness of the major bariatric operations. 4. Understand surgical and nutritional complications of the duodenal switch operation. Introduction Nicola Scopinaro described the original biliopancreatic diversion (BPD) in 1979 in which a distal gastrectomy was performed with an alimentary limb of 250 cm and a common channel of 50 cm [1]. In the original BPD, the blind end of the jejunoileal bypass (JIB) was eliminated. The blind loop was thought to be responsible for bacterial overgrowth and cirrhosis. The BPD created a more substantial stomach pouch of about 250 mL compared to the Roux-en-Y gastric bypass (RYGB) allowing the patient to eat more normal sized meals, and the weight loss was attributed primarily to fat malabsorption. However, the operation was associated with post-­gastrectomy syndromes such as dumping, marginal ulceration, and the potential for diarrhea. The concept of developing the duodenal switch was based on the original studies of DeMeester in dogs that showed when the pylorus was preserved, marginal ulceration was reduced. Hess et al. performed the first BPD/DS in 1988 [2], and Marceau et al. published their results and technique in 1993 [3] in which SG and a post pyloric anastomosis is performed. Ren et al. described the first laparoscopic BPD/DS in 2000 [4], and the R. Sudan (*) Department of Surgery, Division of Metabolic and Weight Loss Surgery, Duke University, Durham, NC, USA e-mail: ranjan.sudan@duke.edu same year Sudan et al. performed the first robotic BPD/DS [5]. The BPD/DS is increasingly being recognized as the most effective bariatric operation for excess weight loss (EWL) and is the best operation for resolution of diabetes and hyperlipidemia. In a propensity matched analysis of different bariatric operations, the comparative effectiveness of BPD/DS showed better odds of resolution of diabetes, hypertension, and more weight loss compared to either the RYGB or the sleeve gastrectomy (SG). It was also better than the SG for resolution of reflux disease but less so than the RYGB [6]. However, BPD/DS annual numbers have lagged behind other bariatric operations [7]. It takes longer to perform, is more challenging technically, and has the potential for more technical complications and nutritional deficiencies which may explain some of the reasons behind its slower adoption. However, with better description of minimally invasive techniques to perform BPD/DS and increasing need to revise RYGB and SG for inadequate results, there is growing interest in performing this more aggressive operation. Preoperative Assessment The indications for performing a BPD/DS are similar to other bariatric operations with preference given to those patients who have a high BMI, more severe diabetes, or hypercholesterolemia. There is evolving evidence that for patients with a BMI of less than 50 kg/m2, the rates of malnutrition or excess weight loss are not any different than those for a BMI greater than 50 kg/m2 [6]. Accordingly, Centers for Medicare and Medicaid allow BPD/DS for patients with BMI > 40 kg/m2 or >35 with a significant comorbidity and sufficient attempt to lose weight through nonsurgical means. Many other payers use similar criteria. The contraindications for performing a BPD/DS are the same as for any bariatric operation such as noncompliance, unresolved psychiatric conditions including substance abuse, or overwhelming medical risk. Any patient in whom malab- © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_14 161 162 sorption should be avoided or those who have anatomical considerations such as dense small bowel adhesions is also not a good candidate for a BPD/DS. Patients are also advised about the side effects of the BPD/DS, and these may be incongruent with their preference or lifestyle. Patients must undergo multidisciplinary evaluations that include medical nutritional and psychological evaluations. The ability to comply with follow-up and with nutritional supplements is even more important with the BPD/DS. Since the operation takes longer to perform, the ability of the patient to tolerate a longer duration of anesthesia is assessed from a cardiopulmonary standpoint. Vitamin deficiencies must be assessed preoperatively and corrected prior to ­surgery as it is often harder to do so after the operation. Patient education regarding appropriate compliance with postoperative followup and adherence to nutritional guidelines is emphasized. In the era of enhanced recovery, a liver shrinking diet may be prescribed, and for the patients who are very heavy, preoperative weight loss may enable completion of the BPD/DS in a single stage. Bowel prep is avoided to prevent dehydration. Preoperatively, the patients are assessed for duration of DVT prophylaxis that is required postoperatively [8]. They are given a high carbohydrate drink in the morning of the surgery as well as prescribed gabapentin and intravenous acetaminophen. A transverse abdominis plane (TAP) block using lipoidal bupivacaine may also be used to reduce the need for postoperative narcotic pain management. Details of the enhanced recovery protocol are available [9]. Technical Details A few years ago, the BPD/DS was primarily being performed by laparotomy on account of its complexity. However, over the last few years, surgeons have become more facile in their minimally invasive technique, and several techniques have been described to perform this operation. The essential details of each technique include a sleeve gastrectomy in which the stomach pouch has a larger capacity than that of a stand-alone primary SG operation and is in the range of 150–250 ml. The pylorus is preserved, and the duodenum is divided where the first part of the duodenum becomes adherent to the pancreas. This often corresponds to the location of the gastroduodenal artery (GDA). A post pyloric anastomosis is performed to the alimentary channel. The technique by which this anastomosis is performed has been described variously using the EEA stapler, linear cutter, handsewn, or robotic. The BPD/DS can be performed in a standard Roux configuration or as a loop duodenoileostomy. Proponents of the duodenoileostomy propose a lowered risk of internal hernias and technical complication due to one less anastomosis between the biliary limb and the alimentary limb, while proponents of the standard configuration think that a leak may manifest with clinically worse signs R. Sudan and symptoms because of the presence of bile. Also, biliary reflux in the single loop version may cause additional longterm problems that have been previously described with Billroth-type operations. This debate is beyond the scope of the present chapter, but it is important to note that there are some variations in the limb length and types of anastomosis of the alimentary channel. In the standard BPD/DS, a 250 cm alimentary limb with 100 cm common channel is commonly used, while the common channel is somewhat longer in the loop configuration and is in the range of 300 cm. In contrast to RYGB, the measurements of the bowel in the BPD/DS are from the ileocecal valve, and the limb lengths are marked using either suture or clips to perform the anastomoses at the appropriate distance and in the correct orientation to prevent twisting of the bowel. Since the average duration of the operation is longer, positioning the patient is important, and they need to be adequately padded and protected to prevent pressure sores. While some surgeons prefer the split-leg position, many others use a standard supine position. Patient positioning, room setup, and trocar placement are optimized depending on the technique for performing the duodenoileostomy. It is important to remember that the operation is performed in three abdominal zones and the room set up accordingly. Access to the abdomen is most often obtained using a Veress needle in the left subcostal area, and an optical trocar is then used to enter the abdominal cavity. Additional ports are placed under direct visualization. The port size needs to consider the size and the brand of the stapler that will be used in the operation. Therefore a liver retractor is placed. The type of liver tractor used is up to the surgeon (Fig. 14.1). Additionally, the liver is often bulky, and occasionally a hiatal hernia repair needs to be performed concomitantly making the liver retractor quite important. Another debatable part of the operation is performing a simultaneous cholecystectomy. Many surgeons believe that gallstone formation is higher with a BPD/DS than the RYGB due to more wasting of bile salts. In the event that a stone slips into the common bile duct, access may be more challenging after a BPD/DS, as there is no remnant stomach. Also, if the patient develops acute cholecystitis, it may be harder to perform a cholecystectomy in the area of the duodenoileostomy due to scarring from the previous operation. However, other surgeons do not want to spend the additional time and prefer to address a cholecystectomy or common bile duct storms postoperatively, as and when they develop. Depending on whether a cholecystectomy is performed or not, most surgeons will begin the actual BPD/DS operation by performing the sleeve gastrectomy. The greater curvature of stomach is mobilized from the left crus of the diaphragm to the junction of the first part of the duodenum and the pancreas. As mentioned previously, it is important to make the stomach pouch larger in the BPD/DS compared to the stand-­ 14 Biliopancreatic Diversion with Duodenal Switch: Technique and Outcomes 163 Fig. 14.2 Marking bowel at predetermined distances (100 cm and 250 cm) from the ilececal valve. (From: Sudan and Podolsky [24]. Reprinted with permission from Springer Nature) Fig. 14.1 Sample port pacement: C camera port, LR liver retractor, 1 and 3 are accessory ports, and usually stapling is performed through port 2 and assistant port (From: Sudan and Podolsky [24]. Reprinted with permission from Springer Nature) alone SG. The mobilization can be performed by any energy device that seals and divides the vessels of the greater curvature reliably. This is often facilitated by entering the lesser sac close to the greater curvature and dividing the leaves of the greater momentum close to the stomach and within the gastroepiploic arcade. The highest short gastric vessel can easily be avulsed, and gentle but adequate retraction to visualize this area is important to prevent bleeding. In addition, it is important to avoid lateral spread of thermal energy to the gastroesophageal junction to avoid leaks in this location. Similar care is used when mobilizing the first part of the duodenum so as to not cause either burns or bleeding. Bleeding obscures visualization and prevents safe transection of the duodenum. A 50–60 French bougie in the stomach is often used as a guide, and the stomach transection is started approximately 5 cm proximal to the pylorus on the antrum. This part of the stomach is thicker than the more proximal stomach. Therefore, a longer leg length stapler is used. Newer staplers are automated and facilitate in selecting the appropriate staple load. More proximally, the stomach becomes thinner, and the leg length can be downsized appropriately to decrease bleeding. Most surgeons reinforce the staple lines on the sleeve to prevent leaks or bleeding. The choice of reinforcement is up to the surgeon, and considerations include cost of the material and the time involved in applying reinforcement. The available choices are buttress material, sutures, or clips. When performing the sleeve gastrectomy, it is important not to narrow the sleeve or spiral the staple line. Appropriate retraction by the assistant and careful application of the stapler prevent these complications. Since the duodenum is thinner, a shorter leg length stapler can be used in this location, and reinforcement is optional. The division of the duodenum is carried out in the area of the GDA, and the surgeon must be careful to not damage this vessel, the portal structures, or the duodenum when performing this maneuver. Familiarity with the anatomy in this area is crucial for safe dissection but not hard to learn with experience. The next part of the operation involves performing the duodenoileostomy at the premarked bowel lengths (Fig. 14.2). The small bowel at the premarked site (250 cm) is brought up for anastomosis to the proximal stapled end of the duodenum (Fig. 14.3). This is typically performed antecolic although retrocolic techniques have also been previously described. The division of the omentum or creation of a transomental or transmesocolic window is performed depending on surgeon technique and tension being placed on the anastomosis. The anastomosis in the standard BPD/DS is performed 250 cm from the ileocecal valve and is longer in the loop duodenal switch version. It is important to maintain the appropriate orientation of the small bowel so as to prevent a twist that results in an internal hernia, or worse, a closed loop obstruction. The method of performing the duodenoileostomy is variable. In the EEA approach, the anvil of a size 21 stapler is passed orally through the pylorus and seated at the stapled line of the proximal duodenum. The EEA stapler is then passed through 164 Fig. 14.3 The sleeve gastrectomy is performed, the duodenum is divided, and the bowel at the 250 cm mark is anastomosed to the proximal stapled edge of the duodenum (From: Sudan and Podolsky [24]. Reprinted with permission from Springer Nature) the small bowel and engaged with the anvil. After the anastomosis has been completed, the enterotomy for the stapler is closed by suture or stapling. This process is more demanding technically than when performing a circular anastomosis for the RYGB because the anvil has to pass through the pylorus and the distal small bowel is often quite narrow. Therefore, some surgeons have adapted the linear stapler approach to perform this anastomosis. They close the enterotomy using another stapler or hand suturing. This stapling technique is faster and relies less on the suturing skills of the surgeon but may not use the full length of the duodenum. A totally handsutured technique can be ­performed either laparoscopically or robotically. While the laparoscopic technique is faster and less expensive, it demands higher skill levels and may not be as comfortable ergonomically as the robotic technique. If the surgeon is planning a loop duodenal switch, the operation is completed by performing a leak test using either air or methylene blue. In the standard duodenal switch, the distal anastomosis is performed next by approximating the bowel at the 100 cm mark to the biliary limb. The technique for performing the ileoileostomy is more standard. Most often, a 60 mm stapler is used, and the enterotomy for the stapler is closed using suturing or stapling. The surgeon must ensure that the bowel is not narrowed in this process. Finally, the biliary limb is divided near the duodenoileostomy to complete the standard Roux configuration of the BPD/DS (Fig. 14.4). In order to prevent internal hernias, the closure of mesenteric defects R. Sudan Fig. 14.4 Completion of standard duodenal switch by performing an anastomosis between the alimentary limb at the 100 cm mark to the biliary limb just proximal to the duodenoileal anastomosis and then dividing the biliary limb (From: Sudan and Podolsky [24]. Reprinted with permission from Springer Nature) between the alimentary limb and the biliary limb using a running nonabsorbable suture has been standard practice. In the antecolic version, and particularly with the loop duodenal switch, the closure of Petersen’s defect is debatable. Proponents of closure argue a reduction in internal hernias due to one less anastomosis, and those who leave it open feel that a wide open space is less likely to obstruct. Closure of this space is more difficult in the BPD/DS. Endoscopy with air insufflation to check for patency, leaks, and bleeding is often performed prior to completion of the operation. Methylene blue can also be passed through an orogastric tube to check for leaks. Specimens are retrieved at the end of the case. Larger port sites are closed with a port closure device, and lipoidal bupivacaine can now be injected for TAP block, if not done preoperative, to help with pain control. Postoperative Management After a complex bariatric operation, patients undergoing the BPD/DS are monitored in a unit with staff that is adequately trained and equipment that is suitable for managing such patients. With greater implementation of enhanced recovery programs in bariatric surgery, the use of narcotic medication and routine use of patient-controlled analgesia or epidural anesthesia have become the exception. However, patients 14 Biliopancreatic Diversion with Duodenal Switch: Technique and Outcomes should still be monitored closely as many suffer from sleep apnea. There are some nuances in managing the diet of BPD/ DS patients that are highlighted here. Patients undergoing a BPD/DS have an intact pylorus and have undergone a sleeve gastrectomy. As a result, it is not unusual for patients to have some degree of gastroparesis or pylorospasm similar to that seen in the stand-alone SG patients. This may predispose them to nausea, and judicious use of anti-nausea medicine, including scopolamine patches, has been very helpful in alleviating this problem. In the RYGB, the stomach pouch is small, and there is no restriction to outflow of fluid from the pouch. Hence, RYGB patients tolerate oral intake sooner than the SG or the BPD/DS patients. The BPD/DS patients also have a larger stomach pouch, and therefore high-volume emesis with subsequent chances of aspiration, needs to be kept in mind when initiating oral intake. Once the patients are nausea-free and are on a normal clinical track, liquids are initiated in small quantities and advanced to protein shakes. The duration of the liquid diet varies with bariatric program but is commonly about 2 weeks, at which time patients are started on a soft diet and gradually progressed to more normal consistency diet, over the next 3 months. Vitamins are supplemented as per ASMBS recommendations for malabsorptive operations [10]. If a leak test has been performed during the operation, routine postoperative upper gastrointestinal series is not performed, unless clinically indicated. Early ambulation with assistance is encouraged, and routine use of drains, nasogastric tubes, and Foley catheter is not usually practiced. For DVT prophylaxis, sequential compression devices are used routinely, and appropriate anticoagulation medication is administered, as assessed by the patient’s need preoperatively. Patients are ready for discharge when they are tolerating adequate amount of liquid to prevent dehydration, pain is under good control, and they are ambulating well. The average length of stay for a BPD/DS patient is longer than that of stand-alone SG or RYGB. When patients have return of bowel function, they may experience urgency and frequency. This normalizes to an average of 2–3 soft bowel movements a day. Patients need to be counseled regarding this expectation. If bowel movements are excessive and an infectious colitis has been ruled out, antidiarrheal agents such as loperamide or diphenoxylate-atropine can be used. Complications Surgical Comparative effectiveness among different bariatric operations shows higher complication profile at 30 days and 1 year for the BPD/DS and is discussed below in greater detail in the section on outcomes [6]. Leaks and bleeding are man- 165 aged similar to other bariatric operations, and the need to manage these complications operatively depends on hemodynamic stability as well as radiographic findings. Evidence of a free leak necessitates an urgent operation. Stenting a sleeve leak or a DI anastomosis leak, draining any fluid collections percutaneously, making the patient nil per os, and feeding them through parenteral access, can avoid an operation in suitable patients. Bleeding can be managed with fluid and blood replacement or endoscopic management using epinephrine injections or clips. More aggressive bleeding or one that cannot be accessed endoscopically needs operative intervention. The chances of PE are also higher after a longer operation. Bowel obstructions in BPD/DS can be particularly dangerous because these patients do not have a remnant stomach. Obstruction in the biliary limb or common channel may therefore manifest itself with the blowout of the duodenum, if not treated in a timely fashion. Abdominal pain should be investigated with a CAT scan, and patients with dilation of the biliary limb should be promptly explored to prevent this from occurring. Typically, the bowel is traced back from the ileocecal valve to ensure appropriate orientation. Nausea and vomiting may also be attributable to strictures of the sleeve, and a technique to widen the stricture by performing a strictureplasty has been described [11]. Another option is to perform a RYGB proximal to the stricture, but this operation is more complex in the face of a previous BPD/DS. Conversion to a RYGB may also be considered if patients have intolerable reflux. Sometimes in the absence of any anatomic abnormality, nausea is of a metabolic nature and improves with time. Resolution of nausea may take months and requires careful monitoring and supplementation of nutrition in the meantime. Despite the statistically higher complication rate, the rate of complications from BPD/DS is still acceptable from a clinical standpoint. Nutritional Complications Duodenal switch patients do not suffer from dumping symptoms, and as a result, they can eat larger portions and tolerate a wider variety of foods. If they are not careful and consume sugars and starches, weight loss may be inadequate. Also, as the bowels are bypassed quite distally, many patients will become lactose intolerant, and most malabsorb fat including the fat-soluble vitamins such as A, D, E, and K. If patients are not mindful of the type of food they eat, they may also have excessive flatulence and steatorrhea. The fat-soluble vitamins need to be supplemented in their water-soluble analogue form and often need to be specially ordered and cost more. Noncompliance with these vitamins may result in deficiency states. Duodenal switch patients are often more predisposed to vitamin D deficiency and a concomitant increase in parathormone level. Deficiencies related to minerals such 166 as iron, copper, zinc, and magnesium can also be seen. In order to detect and adequately treat these vitamin deficiencies, serum levels need to be assessed at least annually, and more frequently if needed. Recommended doses of vitamins and micronutrients are available through ASMBS guidelines [10]. As stated previously, it is important to obtain these levels preoperatively and correct them prior to surgery. Patients who have chronic nausea and vomiting are also predisposed to vitamin B deficiency [12–19]. Another major concern with the BPD/DS is protein deficiencies. Marceau et al. increased the length of the common channel from 50 to 100 cm and described a decrease in the incidence of protein deficiency compared to the Scopinaro BPD. He also demonstrated reduced bowel movements and improved levels of fat-soluble vitamins [20]. It is conceivable that increasing the length of the common channel in the loop version of the duodenal switch may further decrease nutritional deficiencies. BPD/DS shows remarkable stability in maintaining weight loss over a long period of time, but a few patients need to have the operation reversed due to excessive malnutrition or bowel side effects. In experienced centers, about 2% of the patients may need a reversal for these reasons. This can be accomplished by either creating a more proximal side-to-side anastomosis between the biliary and the alimentary limb, or the Roux limb can be disconnected at its junction with the common channel and moved more proximally. Another issue with more distal bypass is the formation of oxalate kidney stones. Normally the oxalates in the gut bind to calcium and are excreted in stool. In BPD/DS due to inadequate binding in the gut, oxalates are absorbed and excreted through the kidneys where stones can form. This can be avoided by consuming supplemental calcium that binds to oxalates and prevents its absorption. Adequate hydration and making the urine more acidic help dissolve oxalate crystals in the urinary tract. Outcomes The vast majority of studies in the literature are single-center retrospective studies from investigators who have extensive experience with BPD/DS [2, 3, 21]. There is one randomized study that compares BPD/DS to RYGB and patients with a BMI greater than 50 kg/m2 [19]. Sudan et al conducted a multi-institutional propensity matched analysis on comparative effectiveness of bariatric operations that included 130,796 subjects, 1,436 patients underwent BPD/ DS and were compared with 5,942 SG and 66,324 RYGB patients. The remaining underwent LAGB. This study demonstrated that BPD/DS had the greatest BMI change at 1 year, followed by RYGB, SG, and AGB, respectively. When using LAGB as a reference, patients undergoing BPD/DS had a BMI reduction of 10.6 units versus 9.3 for R. Sudan RYGB and 5.7 for SG. BPD/DS had the greatest odds for remission for diabetes type II (OR = 5.62, 95% CI: 4.60– 6.88), RYGB (OR = 3.5, 95% CI: 3.39–3.64), and SG (OR = 2.11, 95% CI: 1.92–2.31). Remission of hypertension was also the best after BPD/DS. However, for GERD the best operation was RYGB followed by BPD/DS, and it is noteworthy that the BPD/DS was still better at resolving reflux than SG. In this study, adverse events (AE) and serious adverse events (SAE) at 30 days and 1 year for the unmatched cohort were higher for the BPD/DS compared to the other operations. At 30 days, unmatched AEs were 8.04% for SG, 11.77% for RYGB, and 20.26% for BPD/ DS. At 1 year, the AE rates were 6.5% for AGB, 10.03% for SG, 17.99% for RYGB, and 27.52% for BPD/DS. SAE rates for AGB at 30 days were 0.23% and increased to 0.81% for SG, 1.35% for RYGB, and 3.63% for BPD/DS. At 1 year, SAE rates increased slightly to 0.3% for AGB, 0.93% for SG, 1.58% for RYGB, and 4.60% for BPD/DS [6]. Rates of bleeding at 30 days were 0.63% for SG, 1.38% for RYGB, and 0.99% for BPD/DS. Leaks were 0.14% for SG, 0.36% for RYGB, and 0.89% for BPD/DS. PE was 0.11% for SG, 0.13% for RYGB, and 0.54% for BPD/DS. At 1 year, these rates increased only slightly suggesting that beyond 30 days operative complications remained relatively stable. After matching, the odds of adverse events and severe adverse events remained higher for the BPD/DS at 30 days and 1 year [6]. Longer-term single-center retrospective studies have similarly demonstrated excellent weight loss and resolution of comorbidities. A study comparing BPD/DS with RYGB in patients with a BMI of ≥50 kg/m2 demonstrated faster and better resolution of comorbidities and higher weight loss for BPD/DS with no significant increase in morbidity or mortality [21]. In another study, 810 BPD/DS patients with a BMI < 50 kg/m2 demonstrated excellent weight loss and resolution of comorbid conditions suggesting its appropriateness for lower BMI patients [22]. Patient satisfaction was also excellent with acceptable complications rates. Quality of life studies after BPD/DS are limited with regard to gastrointestinal symptoms. The average BPD/DS patient has been shown to have an average of 2.7 bowel movements a day [23]. Conclusion BPD/DS has better weight loss and resolution of certain comorbid conditions such as diabetes, hypercholesterolemia, and hypertension. The operation is now being performed using a variety of minimally invasive techniques, and although still not very common, its popularity seems to be increasing. It is associated with nutritional and higher technical complication rates that can be managed by an experienced team. 14 Biliopancreatic Diversion with Duodenal Switch: Technique and Outcomes Question Section 1. Regarding the biliopancreatic diversion without the duodenal switch, all of the following are true except: A. There is no blind end. B. A distal gastrectomy is performed. C. The operation has shown excellent weight loss. D. Rate of marginal ulcerations is low. 2. According to a large database analysis comparing Duodenal switch to Roux-en-Y gastric bypass and sleeve gastrectomy operations the following is true: A. Diabetes resolution is the best with a duodenal switch. B. GERD resolution is best with a duodenal switch. C. The operative complications are clinically unacceptably high with a duodenal switch. D. The operative complications are statistically equivalent between a duodenal switch and a gastric bypass. 3. The following is true regarding the duodenal switch: A. Dumping is common. B. Steatorrhea is common. C. Vomiting is common. D. Marginal ulcer is common. 4. Regarding vitamin deficiencies following a duodenal switch, the following is true: A. Vit D deficiency is uncommon. B. Use of water-soluble analogues of fat-soluble vitamin is mandatory. C. Use of fat-soluble analogues of water-soluble vitamins is mandatory. D. Patients do not need supplementation of vitamin K. References 1. Scopinaro N, Gianetta E, Civalleri D, Bonalumi U, Bachi V. Bilio-­ pancreatic bypass for obesity: 1. An experimental study in dogs. Br J Surg. 1979;66(9):613–7. 2. Hess DS, Hess DW. Biliopancreatic diversion with a duodenal switch. Obes Surg. 1998;8(3):267–82. 3. Marceau P, Biron S, Bourque RA, Potvin M, Hould FS, Simard S. Biliopancreatic diversion with a new type of gastrectomy. Obes Surg. 1993;3(1):29–35. 4. Ren CJ, Patterson E, Gagner M. Early results of laparoscopic biliopancreatic diversion with duodenal switch: a case series of 40 consecutive patients. Obes Surg. 2000;10(6):514–23; discussion 24. 5. Sudan R, Puri V, Sudan D. Robotically assisted biliary pancreatic diversion with a duodenal switch: a new technique. Surg Endosc. 2007;21(5):729–33. 6. Sudan R, Maciejewski ML, Wilk AR, Nguyen NT, Ponce J, Morton JM. Comparative effectiveness of primary bariatric operations in the United States. Surg Obes Relat Dis. 2017;13(5): 826–34. 7. English WJ, DeMaria EJ, Brethauer SA, Mattar SG, Rosenthal RJ, Morton JM. American Society for Metabolic and Bariatric Surgery 167 estimation of metabolic and bariatric procedures performed in the United States in 2016. Surg Obes Relat Dis. 2018;14(3):259–63. 8. Aminian A, Andalib A, Khorgami Z, Cetin D, Burguera B, Bartholomew J, et al. Who should get extended thromboprophylaxis after bariatric surgery?: a risk assessment tool to guide indications for post-discharge pharmacoprophylaxis. Ann Surg. 2017;265(1):143–50. 9. Thorell A, MacCormick AD, Awad S, Reynolds N, Roulin D, Demartines N, et al. Guidelines for perioperative care in bariatric surgery: Enhanced Recovery After Surgery (ERAS) society recommendations. World J Surg. 2016;40(9):2065–83. 10. Mechanick JI, Youdim A, Jones DB, Garvey WT, Hurley DL, McMahon MM, et al. Clinical practice guidelines for the perioperative nutritional, metabolic, and nonsurgical support of the bariatric surgery patient – 2013 update: cosponsored by American Association of Clinical Endocrinologists, The Obesity Society, and American Society for Metabolic & Bariatric Surgery. Obesity (Silver Spring). 2013;21(Suppl 1):S1–27. 11. Sudan R, Kasotakis G, Betof A, Wright A. Sleeve gastrectomy strictures: technique for robotic-assisted strictureplasty. Surg Obes Relat Dis. 2010;6(4):434–6. 12. Aasheim ET, Bjorkman S, Sovik TT, Engstrom M, Hanvold SE, Mala T, et al. Vitamin status after bariatric surgery: a randomized study of gastric bypass and duodenal switch. Am J Clin Nutr. 2009;90(1):15–22. 13. Binggeli C, Corti R, Sudano I, Luscher TF, Noll G. Effects of chronic calcium channel blockade on sympathetic nerve activity in hypertension. Hypertension. 2002;39(4):892–6. 14. Bloomberg RD, Fleishman A, Nalle JE, Herron DM, Kini S. Nutritional deficiencies following bariatric surgery: what have we learned? Obes Surg. 2005;15(2):145–54. 15. Dolan K, Hatzifotis M, Newbury L, Lowe N, Fielding G. A clinical and nutritional comparison of biliopancreatic diversion with and without duodenal switch. Ann Surg. 2004;240(1):51–6. 16. Rudnicki SA. Prevention and treatment of peripheral neuropathy after bariatric surgery. Curr Treat Options Neurol. 2010;12(1):29–36. 17. Salle A, Demarsy D, Poirier AL, Lelievre B, Topart P, Guilloteau G, et al. Zinc deficiency: a frequent and underestimated complication after bariatric surgery. Obes Surg. 2010;20(12):1660–70. 18. Slater GH, Ren CJ, Siegel N, Williams T, Barr D, Wolfe B, et al. Serum fat-soluble vitamin deficiency and abnormal calcium metabolism after malabsorptive bariatric surgery. J Gastrointest Surg. 2004;8(1):48–55; discussion 4–5. 19. Sovik TT, Taha O, Aasheim ET, Engstrom M, Kristinsson J, Bjorkman S, et al. Randomized clinical trial of laparoscopic gastric bypass versus laparoscopic duodenal switch for superobesity. Br J Surg. 2010;97(2):160–6. 20. Marceau P, Biron S, Hould FS, Lebel S, Marceau S, Lescelleur O, et al. Duodenal switch improved standard biliopancreatic diversion: a retrospective study. Surg Obes Relat Dis. 2009;5(1):43–7. 21. Prachand VN, Davee RT, Alverdy JC. Duodenal switch provides superior weight loss in the super-obese (BMI > or =50 kg/m2) compared with gastric bypass. Ann Surg. 2006;244(4):611–9. 22. Biertho L, Biron S, Hould FS, Lebel S, Marceau S, Marceau P. Is biliopancreatic diversion with duodenal switch indicated for patients with body mass index <50 kg/m2? Surg Obes Relat Dis. 2010;6(5):508–14. 23. Marceau P, Hould FS, Lebel S, Marceau S, Biron S. Malabsorptive obesity surgery. Surg Clin North Am. 2001;81(5):1113–27. 24. Sudan R, Podolsky E. Totally robot-assisted biliary pancreatic diversion with duodenal switch: single dock technique and technical outcomes. Surg Endosc. 2015;29(1):55–60. Single Anastomosis Duodeno-ileostomy 15 Amit Surve, Daniel Cottam, Hinali Zaveri, and Samuel Cottam Chapter Objectives 1. History of the single anastomosis duodeno-­ ileostomy (SADI) procedure. 2. Describe surgical technique with attention to the steps to prevent common as well as rare complications. 3. Summarize the outcomes in terms of weight loss, complications, nutritional outcomes, and type 2 diabetes mellitus resolution. 4. Describe SADI as a revision procedure following failed sleeve gastrectomy (SG), Roux-en-Y gastric bypass (RYGB), and adjustable gastric banding (AGB). 5. Review of literature. History In 1986, Dr. Douglas Hess modified the biliopancreatic diversion (BPD) in order to retain the pylorus as a functioning part of the digestive system. The new procedure was called a biliopancreatic diversion with duodenal switch (BPD-DS) [1]. This procedure includes removing a significant portion of the stomach as well as bypassing a segment of the small intestine. The new procedure was more effective for long-term weight maintenance than a Roux-en-Y gastric bypass (RYGB) or a vertical banded gastroplasty (VBG). However, it became associated with complications such as chronic diarrhea, smelly stools, flatulence, vitamin deficiencies, and protein-calorie malnutrition. Because of the complication profile, the BPD-DS did not gain widespread adoption. Most surgeons gravitated to the RYGB as the best balance of weight loss and procedural A. Surve · D. Cottam (*) · H. Zaveri · S. Cottam Bariatric Medicine Institute, Salt Lake City, UT, USA related side effects. The RYGB suffered from ulcers, perforations, anastomotic dilation, vitamin and protein deficiencies, dumping syndrome, anastomotic strictures, gastro-gastric fistula, and internal hernias [2]. The complications of the RYGB are almost all associated with the formation of the Roux limb. To eliminate the problems with Roux limb formation, Drs. Torres and Sanchez-Pernaute from Spain introduced a variant of BPD-DS that involved one anastomosis instead of two. They named it “Single anastomosis duodenoileal bypass with sleeve gastrectomy” (SADI-S) [3]. The SADI-S involved preservation of pylorus like BPD-DS but eliminated the Roux limb found in RYGB and BPD-DS. They used a 54-size Fr bougie for sleeve creation with a 200-cm common channel. However, this length was later enlarged to 250 cm to reduce the risk of hypoproteinemia [4]. In order to reduce a prohibitive risk of short bowel syndrome and provide a margin if the bowel is inaccurately measured, Drs. Cottam and Roslin from the United States further modified the SADI-S procedure. Their variant included a 40-size Fr bougie (vs. 54-size Fr bougie) for better weight loss along with a common channel of 300 cm (vs. 250 cm). This variant was called “Stomach intestinal pylorus-­ sparing surgery” (SIPS) [5]. However, the basic concept of preserving the pylorus and having a single anastomosis was the same. The SIPS, being a fairly new concept in 2015, was considered experimental in the United States [6]. However, by 2018, based on clinical knowledge, expert opinion, and published peer-reviewed scientific evidence, the International Federation for the Surgery of Obesity and Metabolic Disorders (IFSO) considered the SADI-S (and its SIPS variant) an established bariatric procedure [7]. In addition, the IFSO proposed a nomenclature for surgery that combines a partial gastrectomy with a single anastomosis between the small bowel regardless of bougie size or common channel length as SADI-S or one anastomosis duodenal switch (OADS). © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_15 169 170 A. Surve et al. Surgical Technique Sleeve Creation Retrograde Tracing and Temporary Tacking With the creation of the SADI-S, the consensus among surgeons who perform this procedure making the sleeve too “tight” is a problem. By tradition, surgeons have not made sleeve’s smaller than 40 F. However, just as there are many ways to create a sleeve, the same applies to the creation of the sleeve during SADI-S. This would include oversewing, The first step of the procedure is to locate the ileocecal valve (Fig. 15.1). Once this is located, the small bowel is traced retrograde to the desired common channel length (this varies greatly between authors and around the world) and tacked up to the gastrocolic omentum (Fig. 15.2). Fig. 15.1 Location of the ileocecal valve Fig. 15.2 Temporary tacking of the ileal loop limb to the gastrocolic omentum of varying lengths 15 Single Anastomosis Duodeno-ileostomy staple line reinforcement, the distance from the pylorus, and treatment of the hiatus (Fig. 15.3). Duodenal Dissection and Transection There are two equally popular techniques for the dissection of the proximal duodenum. The more traditional technique involves starting the dissection 4 cm distal to the pylorus and gently retracting the duodenum taking down attachment between the colon, pancreas, and liver before division. 171 The other equally popular technique relies on adequate blood supply from the right and left gastric vessels on the lesser curve and was popularized by Cottam and Roslin. Once the sleeve is created, they continue taking down the gastroepiploic perforators after the formation of the sleeve until the attachments from the duodenum to the pancreatic head stops easy progress [8]. This is usually 3 cm beyond the pylorus. This technique spares vessels from the pancreaticoduodenal arch from injury but sacrifices the gastroepiploic perforators to do so (Fig. 15.4). A band passer is passed through the space created under the duodenal bulb, toward the liver, and through Fig. 15.3 Sleeve creation with variable sized bougie Fig. 15.4 Important blood supply to the duodenum that needs to be preserved during duodenal dissection and transection 172 Fig. 15.5 Creation of space under duodenal bulb through the gastrohepatic ligament A. Surve et al. nal stump staple line to relieve tension with a running suture (Fig. 15.7). Then enterotomies are made as large as possible and approximately the same size on both the small bowel and the duodenum (Fig. 15.8). A posterior row and an anterior row are sewn closed with running suture. Some surgeons also perform a second anterior row. Other surgeons have perfected a three-stapler technique to transect the duodenum, connect the small bowel, and close the enterotomy. This involves dilating the proximal duodenum using air from the sizing tube. Then enterotomies are made in both the small bowel and proximal duodenum. The duodenum enterotomy is made 1 cm from the pylorus. The stapler is fired on the anterior portion of the duodenum away from the staple line. Next sutures are placed on the proximal and distal ends of the enterotomy and elevated to apply the stapler. Anti-reflux Stitch Some patients can experience nausea when food goes primarily down the afferent limb of the loop. To prevent this rare complication, an anti-reflux stitch is placed (Fig. 15.9). This involves placing an interrupted suture between the afferent limb to the antrum of the stomach to prevent retrograde filling of the afferent limb [9]. The entire SADI-S procedure can be seen in Fig. 15.10. Fig. 15.6 Transection of the duodenum Postoperative Care the gastrohepatic ligament, with care taken to preserve the blood vessels (Fig. 15.5). This technique also results in a linear gastric tube since the gastropancreatic ligaments are responsible for the curve of the stomach, and when you eliminate them, the antrum assumes a linear shape. Once the duodenum is dissected free with either technique, a linear stapler is placed across the first part of the duodenum (Fig. 15.6). Before firing the stapler, always try to feel the pylorus to make sure the stapler is distal to the pylorus since it is not always easy to visualize the pylorus. The care of the SADI-S patient does not differ substantially from the care of the sleeve patient or the gastric bypass patient. Follow-up is similar as well except greater emphasis is placed on the patient’s postoperative supplementation. Current recommendations for postoperative labs are at 6 months, 1 year, and then yearly thereafter. The recommended yearly nutritional evaluation includes folate, ferritin, parathyroid (PTH), vitamins A, D, E, K, B1, and B12, copper, and zinc. Depending on circumstances, we may also measure hemoglobin A1C, complete blood count (CBC), comprehensive metabolic panel (CMP), lipid panel, prealbumin, phosphorus, fasting insulin levels, thyroid-stimulating hormone (TSH), and free T4. Duodeno-ileostomy (DI) Unlike the gastrojejunal anastomosis in the gastric bypass, practitioners of duodenal switch surgery do not believe the duodenal small bowel connection plays any role in short- or long-term weight loss or maintenance. Therefore, the emphasis is not on size but safety. Currently, the most popular technique worldwide is the totally hand-sewn technique. For the hand-sewn technique, the mesenteric border of the loop limb is sewn to the duode- Weight Loss Outcomes with SADI-S Following the procedure, the patients can lose 78–95% of the excess weight at 18 months (weight loss peak) depending on their starting BMI [4, 10–15]. These results seem to be maintained out 4–6 years from surgery [10, 14]. 15 Single Anastomosis Duodeno-ileostomy 173 Fig. 15.7 Approximation of the loop limb to the proximal duodenal stump Fig. 15.8 Creation of the hand-sewn duodeno-ileostomy Complications One of the reasons Drs. Torres, Sanchez-Pernaute, Cottam, and Roslin introduced the SADI-S procedure and its variant was to minimize the complications seen with BPD-DS and RYGB. The primary advantage of SADI over BPD-DS is that there is no distal anastomosis or Roux limb. They postulated that with one anastomosis and no Roux limb, the complications related to 174 Fig. 15.9 Creation of the anti-reflux stitch A. Surve et al. anastomoses could be lowered. In a recent study, Surve et al. specifically studied the incidence of complications related to one anastomosis (loop DI) following 1,328 SADI-S cases as well as compared the anastomotic complication rates to the reported rates following BPD-DS and RYGB in the literature [16]. They found that the anastomotic leak, ulcer, and bile reflux occurred in 0.6%, 0.1%, and 0.1%, respectively. None of the patients experienced volvulus at the DI or an internal hernia. The overall incidence of complications associated with loop DI was far lower than the reported incidence of anastomotic complications following BPD-DS and RYGB. These numbers from the study included the learning curve of each practitioner; so long-term results are expected to be even better than those reported in the study. The most common short- and long-term complications one can expect are nausea (2.2%), wound infection (2.2%), and sleeve stricture (2.9%), respectively [10, 12]. However, Fig. 15.10 Hand-drawn sketch of a single anastomosis duodeno-ileostomy with sleeve gastrectomy procedure along with an upper gastrointestional series 15 Single Anastomosis Duodeno-ileostomy these complications are not explicitly related to the SADI-S procedure. The most common complication related to the SADI-S procedure is chronic diarrhea that is seen in approximately 2% patients [10]. Not all the patients that experience chronic diarrhea require limb lengthening. In some patients, it can be treated with dietary manipulation, probiotics, and Lomotil as well. With 300 cm approximately, 1% of the patients will need limb lengthening over a 5-year period [10]. The overall short- and long-term complication rates seen following SADI-S were 7.7% and 10.9%, respectively [10]. Rare Complications and Their Management Retrograde Filling of Afferent Limb So far there have been only two cases reported in the literature [9]. In both cases, scar tissue was found distal to the DI that pulled the efferent limb superior to the DI, causing the flow of food and secretions down the afferent limb. Tacking the afferent limb to the antrum of the stomach hopefully can prevent such complication. Miscounted Bowel Miscounted common channel could be a part of the learning curve. Usually, such patients present with chronic diarrhea and are usually treated by lengthening the common channel. The length of the additional common channel depends on the miscounted bowel. The symptoms are usually resolved following this procedure [17]. Reversed Loop The reversed loop is another rare complication that can occur as a part of learning curve. The patients usually present with persistent nausea and vomiting. The ideal way to fix this complication is redoing the duodenal small bowel anastomosis [10]. Nutritional Outcomes Contrary to popular belief, there is minimal nutritional malabsorption seen following the SADI-S procedure, and no author has presented evidence of primary malnutrition with 250–300 cm of small bowel for absorption [18]. Typically, in a patient with a 300-cm common channel, 65% fat malabsorption can be seen. Even though the percent- 175 age of fat malabsorption with SADI-S is higher when compared to the RYGB procedure [11], with the recommended postoperative multivitamin regimen, the patients usually do not experience fat-soluble vitamin deficiency. Zaveri et al. in the 4-year outcome paper on SADI-S studied the fat-soluble vitamin levels, pre- and postoperatively [10]. At 4 years, even though 7.5% patients had a vitamin A deficiency, there was no statistically significant difference when compared to the preoperative abnormal levels. Prevalence of vitamin D deficiency is reported to be as high as 90% in patients with obesity. In their study, 48.2% had vitamin D deficiency preoperatively. However, at 4 years, only 23% patients experienced vitamin D deficiency. This shows that despite the shorter bowel lengths, vitamins can still be replete. Vitamins E and K are fat-soluble but are far easier to absorb with any vitamin regimen. At 1 year in a recent study, no patients experienced vitamins E and K deficiency [10]. The nutritional deficiencies that are reported in the literature can be seen in Table 15.1 [4, 11, 13–15, 19–23]. To conclude, the SADI-S procedure is not associated with apparent nutritional changes; however, postoperative supplementation with iron, multivitamins, calcium, and vitamin D may be required continuously to prevent nutritional deficiency. T2DM Resolution The SADI-S procedure is a variant of the DS procedure. As such, it retains the expected long-term diabetes mellitus (DM) resolution seen in DM. Sanchez-Pernaute et al. studied the effect of SADI-S on patients with obesity and DM. Absolute remission for patients with oral therapy was seen in 92.5% in the 1st year and 75% in the 5th year [14], while absolute remission for patients under insulin therapy was seen in 47% in the 1st year and 38.4% in the 5th year. Overall remission rate (HbA1c <6%) was seen in 71.6%, 77%, 75.8%, 63.3%, and 52% at one-fifth year, respectively. A short DM history and no requirement for insulin therapy were related to higher remission rate, while Zaveri et al. found the complete remission rate (HbA1c < 6% without antidiabetic medication) of 78.6%, 77.8%, 81.3%, and 81.3% after one-fourth year, respectively [10]. Similarly, 97.6% of the patients were able to maintain HbA1c < 6% with or without the medication at 4 years. Cottam et al. compared the DM resolution rate at differing levels of HbA1c between RYGB and SADI-S [12]. At each differing level, SADI-S had a statistically higher amount of diabetic resolution compared to RYGB. At 1 year, HbA1c < 6% was controlled in 88% of SADI-S patients versus 58% of RYGB patients without any antidiabetic medications. 8 16 29 8 10 7.6 13 8 3 1 1 1 1 2 Protein Def (%) 1 2 Post-op year 11 8 3.1 12 8 12 13 6 Albumin Def (%) – 5 5.1 – 18 6 13 0 Calcium Def (%) – – – – – 54 – 42 PTH Def (%) – – 45 – – 53 31 40.5 10.8 29 34 23 Vitamin def (%) A D – 46 – 6 SADI-S single anastomosis duodeno-ileostomy with sleeve gastrectomy, Def deficiency, Zn zinc, Cu copper, Fe iron, Se selenium Nelson (2016) [21] Sanchez-Pernaute (2013) [4] Sanchez-Pernaute (2015) [14] Cottam (2016) [11] Cottam (2017) [15] Enochs (2015) [22] Neichoy (2016) [23] Surve (2017) [13] Author Table 15.1 Published nutritional deficiencies following SADI-S – – – – – 7 – E – – – – – – – K 29 38 15.3 10.4 0 8 B12 0 20 16 – – – – 3 Folate – 10 – – – – – 33 – – – – – 11 – – – – – – Mineral def (%) Zn Cu Fe – – – – – 4 – – – – – 28 Se – 2.5 176 A. Surve et al. 15 Single Anastomosis Duodeno-ileostomy The overall T2DM remission rates following SADI-S can be around 81–90% without antidiabetic medication [10, 14, 23]. SADI as Revision Procedure SADI Following Failed SG Weight loss failure and weight recidivism over the long term have been a common concern following SG. Moreover, SG has also shown poor results in patients with BMI >50 kg/m2 [24]. This is one of the reasons why revision procedures following failed SG have been increasing in recent years. The question that remains for the bariatric surgeons is what revision options are now available for patients who fail SG? The SADI-S procedure is increasingly becoming popular as a second step revision procedure or as a staged procedure, primarily for weight loss failure following SG. The mean EWL at 1 and 2 years following the second step SADI has been around 69% and 72%, respectively [25]. More importantly, postoperative complications have been minimal with no reported incidence of small bowel obstruction or internal hernias, which are commonly seen following RYGB and BPD-DS. Zaveri et al. showed that if SADI is performed within the 1st year of performing SG, the patients could lose a similar amount of weight as a primary SADI-S procedure [26]. However, this approach will need a longer follow-up to understand its limitation. 177 Although the data are limited, the initial data have shown to have favorable outcomes. The EWL following revision SADI-S for failed RYGB at 1 year has been reported between 53% and 61% and at 2 years has been reported around 55% [31]. This is similar to patients who have an RYGB to traditional DS [32]. ADI-S Following Failed Adjustable Gastric S Band Multiple studies have demonstrated a high incidence of weight recidivism and long-term complications with AGB [33–35]. In addition, multiple reports have been published on the different approach to revising the failed AGB patients to SG, RYGB, or BPD-DS. However, EWL has not been that favorable, and complications have been high [36–38]. Therefore, the controversy exists regarding the choice for patients with failed LAGB. The outcomes of SADI-S as a revision option following failed AGB have been only reported by Surve et al. in the literature [39]. Technically, it is simpler than BPD-DS and RYGB. Three years report showed 90% EWL following revision of AGB to SADI-S. The weight loss mirrors the weight loss seen following primary SADI-S procedure. The only caveat with this procedure is careful dissection to avoid leaks and strictures from scar tissue below the band. Review of Literature SADI-S Following Failed RYGB The RYGB has been associated with long-term good weight loss outcomes. However, approximately 25% of patients have weight recidivism [2, 27]. Various techniques such as resizing the pouch or lengthening the Roux limb have been used but with limited or no success [28, 29]. The SADI-S is an effective option for failed RYGB patients due to its pyloric preserving capacity. Most of the patients who fail RYGB usually eat small, frequent, high-carbohydrate meals. This is a physiologic response to vacillating blood sugar levels causing hunger [30]. The SADI-S with its pylorus preserving capacity plays an important role in maintaining blood sugar levels and satiety. Although technically possible this revision is challenging and has been shown to have high complication rates [31]. The outcomes with primary and revision SADI/SADI-S that have been published in the literature are summarized in Table 15.2. Unique data sets have been included [4, 5, 10, 18, 21, 23, 25, 31, 39–43]. Conclusion Effective weight loss, high T2DM resolution rates, low anastomotic complication rates compared to other established procedures, minimal nutritional changes, effective second stage, or revision option following failed bariatric procedures make SADI/SADI-S one of the best options for patients with morbid obesity and coexisting conditions. 178 A. Surve et al. Table 15.2 Published outcomes with primary and revision SADI/ SADI-S Article Procedure Primary SADI-S procedure SIPS Mitzman et al. [5] SADI-S Zaveri et al. [10] SADI-S Sanchez-­ Pernaute et al. [4] Neichoy et al. [23] Morales et al. [40] Abd-Elatif et al. [18] Gebelli et al. [41] Nelson l et al. [21] SIPS SESDID SADI-S SADI-S SADI-S Bougie size and Sample common channel size Follow-up 42 F and 300 cm 40 F and 300 cm 54 F and 200 cm (50 patients) 250 cm (50 patients) 40 F and 300 cm 34 F and 300 cm 36 F and 250 cm 36 F and 300 cm 34 F and 250 cm SADI procedure following failed SG SADI 42–54 F and Sanchez-­ 250 cm Pernaute et al. [25] Vilallonga et al. [42] Balibrea et al. [43] SADI SADI Bougie size is unk and 300 cm 32 F and 200–300 cm SADI procedure following failed RYGB SADS 40 F and Surve et al. [31] 300 cm SADI procedure following failed AGB SIPS 40 F and Surve et al. [39] 300 cm Weight loss T2DM Rem. rate Complication Fat-soluble vitamin deficiency 123 1 year 72.3%EWL N/A 6.5% N/A 437 4 years 85.7%EWL 81.3% 100 3 years >95% EWL >90% Early = 7.7% Late = 10.9% 5–9% Vit A = 7.5% (4 years) Vit K = 5.8% (1 year) Vit D = 6% Def 40% Ins 225 2 years 88.7% EWL 88.8% N/A N/A Early = 4.8% Late = 8% 12% 100 N/A N/A 37 1 year 67 1–1.5 years 23 kg/m2 BMI red N/A N/A 2.7% N/A N/A 12.8% N/A 69 1 year 61.6% EWL 50% Rem, 33.3% Imp 10.1% Vit D = 45.8% 16 2 years 32.5% EWL (SADI) 72% EWL (SADI+ SG) N/A 88% 6.2% Vit A = 25%, Vit D = 6% Def, 50 Ins, Vit E = 0% 3 3–9 months 33.3% Imp 71.4% 0% N/A 30 2 years Early = 13.3% Vit D = 55.5% 44.2%EWL (SADI) 78.9% EWL (SADI+ SG) N/A Late = 50% 23 2 years 54.5% EWL N/A Early = 17.3% Late = 13% Vit D = 9% 27 3 years 90% EWL 75% Early = 40.7% Late = 25.9% Vit A = 0% Vit D = 41.1%, Vit E = 5.8%, Vit K = 0% SESDID Single end-to-end anastomosis duodenoileal, Def deficiency, Ins insufficiency, mos months, Rem remission, Imp improvement, DIOS distal loop duodeno-ileostomy, SADS single anastomosis duodenal switch, SIPS stomach intestinal pylorus-sparing surgery, SG sleeve gastrectomy, RYGB Roux-en-Y gastric bypass, AGB adjustable gastric banding Question Section 1. Which of these is false about SADI surgery? A. Technically it is more difficult than BPD-DS. B. There is an alimentary as well as biliopancreatic limb involved. C. Conversion of failed RYGB to SADI is a feasible option. D. It has fewer ulcers than BPD-DS. 2. One of the main advantages of SADI over RYGB A. Nutritional deficiencies B. Weight loss C. Dumping syndrome D. Protein-energy malnutrition 3. The weight loss between SADI-S with a 300-cm common channel and BPD-DS with a 150-cm common channel and 150 cm Roux limb is A. The same amount of weight loss between the two B. Less weight loss with SADI-S C. Much better weight loss with SADI-S D. Much worse weight loss with SADI-S 15 Single Anastomosis Duodeno-ileostomy 4. What is the percentage of bile reflux seen following SADI surgery in the literature? A. 0.1% B. 1% C. 0.8% D. 2% 179 16. Surve A, Cottam D, Sanchez-Pernaute A, et al. The incidence of complications associated with loop duodeno-ileostomy after single anastomosis duodenal switch procedures among 1328 patients: a multicenter experience. Surg Obes Relat Dis. 2018;14:594–601. 17. Surve A, Zaveri H, Cottam D. A step-by-step surgical technique video with two reported cases of common channel lengthening in patients with previous stomach intestinal pylorus sparing surgery to treat chronic diarrhea. Surg Obes Relat Dis. 2017;13(4):706–9. 18. Abd-Elatif A, Yousser T, Farid M, et al. Nutritional markers after loop duodenal switch (SADI-S) for morbid obesity: a technique with favorable nutritional outcome. J Obes Weight Loss Ther. References 2015;5(3):1000268. 19. Topart P, Becouarn G. The single anastomosis duodenal switch modification: a review of the current literature on outcomes. Surg 1. Hess DS, Hess DW. Biliopancreatic diversion with a duodenal Obes Relat Dis. 2017;12:1306–12. switch. Obes Surg. 1998;8:267–82. 2. Zaveri H, Dallal RM, Cottam D, et al. Indications and operative out- 20. Shoar S, Poliakin l, Rubenstein R, Saber AA. Single anastomosis duodenoileal switch (SADIS): a systemic review of efficacy and comes of gastric bypass reversal. Obes Surg. 2016;26(10):2285–90. safety. Obes Surg. 2018;28(1):104–13. 3. Sánchez-Pernaute A, Rubio Herrera MA, Pérez-Aguirre E, et al. Proximal duodenal-ileal end-to-side bypass with sleeve gastrec- 21. Nelson L, Moon RC, Teixeira AF, Galvao M, Ramos A, Jawad MA. Safety and effectiveness of single anastomosis duodenal tomy: proposed technique. Obes Surg. 2007;17(12):1614–8. switch procedure: preliminary result from a single institution. Arg 4. Sánchez-Pernaute A, Rubio MÁ, Pérez Aguirre E, Barabash A, Bras Cir Dig. 2016;29(Suppl 1):80–4. Cabrerizo L, Torres A. Single-anastomosis duodenoileal bypass with sleeve gastrectomy: metabolic improvement and weight loss 22. Enochs P. The laparoscopic stomach, intestinal and pylorus sparing (SIPS) procedure: a single center analysis of our first 100patients. in first 100 patients. Surg Obes Relat Dis. 2013;9(5):731–5. Surg Obes Relat Dis. 2015;11(6):S165–6. 5. Mitzman B, Cottam D, Goriparthi R, et al. Stomach intestinal pylorus sparing (SIPS) surgery for morbid obesity: retro- 23. Neichoy BT, Schniederjan B, Cottam DR, et al. Stomach Intestinal Pylorus-Sparing Surgery for morbid obesity. JSLS. 2018;22(1). pii: spective analyses of our preliminary experience. Obes Surg. e2017.00063. 2016;26(9):2098–104. 6. Kim J, American Society for Metabolic and Bariatric Surgery 24. Brethauer SA, Hammel J, Schauer PR. Systematic review of sleeve gastrectomy as a staging and primary bariatric operation. Surg Obes Clinical Issues Committee. ASMBS statement on single-­ Relat Dis. 2009;5:469–75. anastomosis duodenal switch. Surg Obes Relat Dis. 2016;12:944–5. 7. Brown WA, Ooi G, Higa K, Himpens J, Torres A, on behalf of the 25. Sánchez-Pernaute A, Rubio MA, Conde M, et al. Single-­ anastomosis duodenoileal bypass as a second step after sleeve gasIFSO-appointed task force reviewing the literature on SADI-S/ trectomy. Surg Obes Relat Dis. 2015;11(2):351–5. OADS. Single anastomosis duodenal-ileal bypass with sleeve gastrectomy/one anastomosis duodenal switch (SADI-S/OADS) IFSO 26. Cottam D, Surve A, Zaveri H, Cottam A. SIPS is quickly becoming an option for surgical revision of failed sleeve gastrectomy. position statement. Obes Surg. 2018;28:1207–16. Bariatric Times. 2016;13(11 Suppl B):B23–4. 8. Surve A, Zaveri H, Cottam D. A safer and simpler technique of duodenal dissection and transection of the duodenal bulb for duo- 27. Dykstra MA, Switzer NJ, Sherman V, Karmali S, Birch DW. Roux en Y gastric bypass: how and why it fails? Surgery Curr Res. denal switch. Surg Obes Relat Dis. 2016;12(4):923–4. 2014;4:165. 9. Surve A, Zaveri H, Cottam D. Retrograde filling of the afferent limb as a cause of chronic nausea after single anastomosis loop duodenal 28. Kaminski DL. Gastric restrictive procedures to treat obesity: reasons for failure and long-term evaluation of the results of operative switch. Surg Obes Relat Dis. 2016;12(4):e39–42. revision. Int J Surg Investig. 2001;2(5):413–21. 10. Zaveri H, Surve A, Cottam D, et al. Mid-term 4-year outcomes with single anastomosis duodenal-ileal bypass with sleeve gastrectomy 29. Sugerman HJ, Kellum JM, DeMaria EJ. Conversion of proximal to distal gastric bypass for failed gastric bypass for superobesity. J surgery at a single US center. Obes Surg. 2018;28:3062–72. Gastrointest Surg. 1997;1(6):524–6. 11. Cottam A, Cottam D, Roslin M, et al. A matched cohort analysis of sleeve gastrectomy with and without 300 cm loop duodenal switch 30. DeMaria EJ. Bariatric surgery for morbid obesity. N Engl J Med. 2007;356:2176–83. with 18-month follow-up. Obes Surg. 2016;26(10):2363–9. 12. Cottam A, Cottam D, Zaveri H, et al. An analysis of mid-term com- 31. Surve A, Zaveri H, Cottam D, Belnap L, Medlin W, Cottam A. Mid-­ term outcomes of gastric bypass weight loss failure to duodenal plications, weight loss, and type 2 diabetes resolution of Stomach switch. Surg Obes Relat Dis. 2016;12(9):1663–70. Intestinal Pylorus-Sparing Surgery (SIPS) versus Roux-En-Y Gastric Bypass (RYGB) with three-year follow-up. Obes Surg. 32. Parikh M, Pomp A, Gagner M. Laparoscopic conversion of failed gastric bypass to duodenal switch: technical considerations and 2018;28:2894–902. preliminary outcomes. Surg Obes Relat Dis. 2007;3(6):611–8. 13. Surve A, Zaveri H, Cottam D, et al. A retrospective comparison of biliopancreatic diversion with duodenal switch with single anas- 33. Himpens J, Cadière GB, Bazi M, Vouche M, Cadière B, Dapri G. Long-term outcomes of laparoscopic adjustable gastric banding. tomosis duodenal switch (SIPS-stomach intestinal pylorus sparing Arch Surg. 2011;146(7):802–7. surgery) at a single institution with two year follow-up. Surg Obes 34. Arapis K, Tammaro P, Parenti LR, et al. Long-term results after Relat Dis. 2017;13(3):415–22. laparoscopic adjustable gastric banding for morbid obesity: 18-year 14. Sanchez-Pernaute A, Rubio MA, Cabrerizo L, et al. Single follow-up in a single university unit. Obes Surg. 2017;27(3):630–40. anastomosis duodenoileal bypass with sleeve gastrectomy (SADIS) for obese diabetic patients. Surg Obes Relat Dis. 35. Victorzon M, Tolonen P. Mean fourteen-year, 100% follow-up of laparoscopic adjustable gastric banding for morbid obesity. Surg 2015;11(5):1092–8. Obes Relat Dis. 2013;9(5):753–7. 15. Cottam A, Cottam D, Portenier D, et al. A matched cohort analysis of Stomach Intestinal Pylorus Saving (SIPS) surgery versus bilio- 36. Carr WR, Jennings NA, Boyle M, et al. A retrospective comparison of early results of conversion of failed gastric banding to sleeve gaspancreatic diversion with duodenal switch with two-year follow-up. trectomy or gastric bypass. Surg Obes Relat Dis. 2015;11(2):379–84. Obes Surg. 2017;27(2):454–61. 180 37. Rutledge R. Revision of failed gastric banding to mini-gastric bypass. Obes Surg. 2006;16(4):521–3. 38. Poyck PP, Polat F, Gouma DJ, et al. Is biliopancreatic diversion with duodenal switch a solution for patients after laparoscopic gastric banding failure? Surg Obes Relat Dis. 2012;8(4):393–9. 39. Surve A, Zaveri H, Cottam D, et al. Laparoscopic stomach intestinal pylorus-sparing surgery as a revisional option after failed adjustable gastric banding: a report of 27 cases with 36-month follow-up. Surg Obes Relat Dis. 2018;14:1139–48. 40. Morales H, Berger F, Espinoza M, Anthon B. Gastric sleeve and duodenal-ileal end-to-side diversion in bariatrics: experience in 100 cases. BMI. 2012;2.3.2 (76–79). A. Surve et al. 41. Gebelli JP, Gordejuela AG, Ramos AC, et al. SADI-S with right gastric artery ligation: technical systematization and early results. Arq Bras Cir Dig. 2016;29(Suppl 1(Suppl 1)):85–90. 42. Villalonga R, Fort JM, Caubet E, et al. Robotically assisted single anastomosis duodenoileal bypass after previous sleeve gastrectomy implementing high valuable technology for complex procedures. J Obes. 2015;2015:586419. 43. Balibrea JM, Vilallonga, Hidalgo M, et al. Mid-term results and responsiveness predictors after two-step single-­ anastomosis duodeno-ileal bypass with sleeve gastrectomy. Obes Surg. 2017;27(5):1302–8. Laparoscopic One Anastomosis Gastric Bypass: History of the Procedure Surgical Technique and Outcomes 16 Helmuth T. Billy, Moataz M. Bashah, and Ryan Fairley Chapter Objectives 1. Familiarize the reader with the evolution of one anastomosis gastric bypass. 2. Present two common techniques described when performing one anastomosis gastric bypass. 3. The reader will understand the controversial issue of bile reflux as well as other complications, how to diagnosis these conditions, and therapeutic options for treatment. 4. Compare and understand the benefits of one anastomosis procedures versus two anastomosis bypass with Roux reconstruction. History In 2001, an article was published in the journal Obesity Surgery reporting operative technique and outcomes in a series of 1274 cases utilizing a single anastomosis version of a gastric bypass [1]. The procedure described a simplified version of a traditional Roux-Y gastric bypass, avoided creation of a small gastric pouch or permanent gastric resection, and eliminated the second distal anastomosis which was required for the Roux-Y gastric bypass or duodenal switch. The concept of anastomosing a loop of jejunum to a long, narrow, gastric pouch created 2–3 cm below the crow’s foot H. T. Billy (*) Metabolic and Bariatric Surgery, Community Memorial Hospital, Ventura, St. John’s Regional Medical Center, Oxnard, CA, USA M. M. Bashah Division of Metabolic and Bariatric Surgery, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar R. Fairley Surgical Education, Community Memorial Hospital, Ventura, CA, USA along the lesser curve was first introduced by Rutledge. Rutledge coined the phrase “mini gastric bypass” (MGB) and described a less complicated operative strategy that was easier to perform and eliminated the need for a Roux limb. It was hypothesized that a one anastomosis approach to gastric bypass could obtain similar results as the Roux-Y gastric bypass while also reducing the side effects and complications associated with the creation of the Roux limb necessary in RYGB and duodenal switch. Attempts at introducing a loop gastric bypass as a bariatric operation had been introduced by Mason 30 years earlier [2]. The “mini gastric bypass” operation introduced by Rutledge differed quite significantly from the original loop gastric bypass described by Mason. Mason’s attempt to create a weight loss operation utilizing a horizontal gastroplasty and a simple loop jejunostomy was based very closely on the Billroth II gastrojejunostomy. Despite the simplicity of the operation, the initial loop gastric bypass created by Mason resulted in uncontrolled bile reflux and exposure of the distal esophagus to the deleterious effects of bile and acid. The controversy created by the esophageal complications following the introduction of the Mason loop gastric bypass has persisted over the two decades since the introduction of the “mini gastric bypass” by Rutledge. Mason’s operation and the reflux producing side effects were quickly abandoned due to the severe and excessive presence of bile reflux on the distal esophagus. Subsequently, the potentially harmful effects of combining bile with acid and exposing the distal esophagus to this combination were well described by DeMeester [3, 4]. Fifty years after Mason first described his looped gastric bypass, the concerns that biliopancreatic reflux reaching the distal esophagus from the afferent limb of a loop gastrojejunostomy persist. Fear that bile induced esophageal inflammation, Barrett’s changes, and esophageal cancer has limited adoption of one anastomosis gastric bypass (OAGB) in the United States. In the 20 years since the mini gastric bypass and OAGB procedures were described, there have been no publications definitively linking these single anastomosis procedures with the development of any cases of esophageal © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_16 181 182 or gastric cancer. On the other hand, reflux secondary to sleeve gastrectomy may contribute to development of Barrett’s esophagus in as much as 17% of patients undergoing vertical sleeve gastrectomy [5]. It has been hypothesized that the low-pressure environment of gastric bypass procedures and the longer length of the gastric pouch in OAGB operations may play a protective role in limiting reflux episodes from reaching the distal esophagus in patients undergoing OAGB. While single anastomosis gastric bypass has been slow to gain traction in the United States, the opposite has been true in Europe, Asia, and the Middle East. Since 2001, over 16,000 patients have been reported in publications reporting OAGB outcomes [6]. The most comprehensive literature review identifying studies reporting outcomes following ­single anastomosis gastric bypass procedures was recently published as an IFSO position statement on March 29, 2018 [7, 8]. Included in the IFSO position statement was the recommendation that the “One Anastomosis Gastric Bypass is a recognized bariatric/metabolic procedure and should not be considered investigational.” The IFSO Task force contributing to the review was composed of a multinational group of 22 recognized and accomplished bariatric surgeons. The United States was represented by two former ASMBS presidents. The task force reviewed, summarized, and correlated the findings of 52 studies reporting outcomes of 16,546 patients to report on the safety and efficacy of one anastomosis gastric bypass procedures. H. T. Billy et al. staged approach [9, 10]. Reversal of a single anastomosis gastric bypass is also relatively and technically easy should it be necessary in the future. OAGB is a simpler, less costly operation to perform. Shorter operative times, less postoperative pain, decreased nausea and vomiting, early ambulation, and little more than an overnight hospitalization have been reported in comparative studies evaluating OAGB to RYGB [11]. The simplification in operative technique allows an inpatient hospitalization for RYGB to become an outpatient procedure for OAGB. The enteroenterostomy in RYGB can require up to four additional staple cartridges and closure of three mesenteric defects. OAGB is a procedure that can be offered by facilities at a significantly reduced overhead cost than that associated with gastric bypass. The improved economics associated with OAGB can significantly increase access to care for those patients who do not have the insurance coverage or economic means to afford more expensive procedures. The widespread adoption of this less expensive, less complex, and possibly safer alternative than RYGB has allowed bariatric surgery to be offered in large numbers to patients who might otherwise not have access to bariatric procedures across Asia, the Middle East, and Europe. There is neither data nor established consensus as to an ideal or recommended operative technique. The initial operation described by Rutledge (MGB) and used in his 2001 series describes a gastric pouch based on the lesser curve of the stomach [12]. The initial staple line is created 1–2 cm below the crow’s foot followed by a vertical extension along the body of the stomach and completing the pouch by stayIntroduction ing just lateral to the angle of His as the stomach is transected. The anastomosis between the pouch and the jejunum One anastomosis gastric bypass (OAGB) is a technically is a wide, end to side anastomosis utilizing the entire length easier operation than Roux-Y gastric bypass or biliopancre- of a 45 mm stapler and is created between 180 and 220 cm atic diversion with duodenal switch. The hallmark of the distal to the ligament of Treitz. There is also no consensus on OAGB is the elimination of the Roux Limb. The well-­ the optimal length of the afferent loop. Variations in the recognized complications associated with the creation of the length of the afferent limb have been described for the RYGB enteroenterostomy are eliminated. Morbidity and elderly, vegetarian patients, and diabetics [13]. Fluctuations mortality rates are lower due to simplified operative dissec- in the distance from the gastrojejunostomy to the ligament of tion. Both the duodenal switch and the Roux-Y gastric Treitz have been reported from 180 to about 250 cm to bypass require the creation of a second enteroenterostomy. greater than 300 cm depending on the age, eating habits, and Well-known complications following RYGB can occur at the comorbid conditions of the patient (Fig. 16.1). enteroenterostomy and include internal anastomotic staple An alteration to the original procedure described by line bleeding, intussusception, internal hernia, and leakage Rutledge using a side-to-side (lateral to lateral) anastomosis from a second anastomosis. These complications are avoided between the jejunal loop and the gastric pouch was described completely with the OAGB. OAGB offers reductions in by Carbajo in 2001 [14]. The modification was designed speoperative time and less aggressive liver retraction. Division cifically to allow the afferent limb to experience less bile of small bowel mesentery is unnecessary in OAGB. The reflux into the gastric pouch. Carbajo and Caballero described preservation of small bowel mesenteric blood flow may the operation as a “one anastomosis gastric bypass” (OAGB). result in a decrease in the incidence of DVT, portal vein Their technique utilized this variation of the anastomosis thrombosis, and bleeding. Large, super-morbidly obese described by Rutledge and lengthened the distance between patients whose BMIs exceed 50 can undergo OAGB without ligament of Treitz and the gastrojejunostomy to between 250 the need for a longer operation or one that would require a and 350 cm (Fig. 16.2). 16 Laparoscopic One Anastomosis Gastric Bypass: History of the Procedure Surgical Technique and Outcomes Fig. 16.1 Technique of the “mini gastric bypass” as described by Rutledge consists of a long conduit from below the crow’s foot extending up to the left of the angle of His. The operation has a wide gastrojejunal anastomosis to an anti-colic loop of jejunum 150–200 cm distal to the ligament of Treitz. The gastrojejunostomy between the posterior wall of the gastric pouch and the antimesenteric border of the jejunum is typically stapled using a 45 mm stapler in an end-to-side-type configuration Since 2001, the variations in technique have resulted in multiple different versions of essentially the same operation [15]. These operations can be found in the medical literature described as: 1. Mini gastric bypass (MGB) 2. One anastomosis gastric bypass (OAGB) 3. Omega loop gastric bypass (OLGB) 4. Single anastomosis gastric bypass (SAGB) This chapter will present the basic construction, operative technique complications, and outcomes we have employed and experienced with the introduction of OAGB into our practice. There is as yet no “gold standard” method with which to perform this procedure. Much of the operative ­techniques preferred by individual surgeons are based on opinions and personal experience. Anastomotic techniques vary between practices, and there are no comparative studies evaluating these differences in operative technique. Variations in technique regarding pouch size, bougie size, gastrojejunostomy technique, or afferent limb length among surgeons 183 Fig. 16.2 “One anastomosis gastric bypass” procedure as described by Carbajo [14] utilizes a side-to-side configuration and a 5–6-cm-long “anti-reflux” suture to approximate the mesenteric border of the jejunal loop to the lateral wall of the gastric pouch. Following this configuration, an enterotomy is made in the distal gastric pouch and the antimesenteric border of the jejunal loop. A stapled gastrojejunostomy in a side-to-side configuration completes the anastomosis performing OAGB are common. The most common consensus agreement between various authors appears to be a long narrow gastric tube which extends to at or below the crow’s foot of the lesser curve [16]. As surgical innovation continues, new operations that are cheaper, safe, and effective are the drivers that will improve access to bariatric surgery. Participation in national and international registries will allow for the collection of long-­ term data, outcome, and complication analysis. Analysis of the outcomes data is essential to the further advancement in the understanding of new and innovative metabolic and bariatric procedures. As we come to develop a consensus on procedures such as the OAGB, we can expect that future improvement in insurance coverage may ultimately benefit access to care nationally. Patient Selection and Preparation The 1991 National Institutes of Health Consensus Development Conference Statement on Gastrointestinal Surgery for Severe Obesity established guidelines for identifying appropriate candidates for metabolic and bariatric sur- 184 gery operations [17]. Although these guidelines are more than 20 years old, they remain a foundation of bariatric surgery patient selection despite their likely outdated and historical significance. Evaluations of patients for possible MGB-OAGB, in our opinion, require utilization of a multidisciplinary team providing medical, surgical, psychiatric, and nutritional expertise. Evaluation of potential surgical candidates as per NIH guidelines is recommended and allows for appropriate discussions of both surgical and nonsurgical approaches to weight loss operations. In 2004, the American Society for Metabolic and Bariatric Surgery published a Consensus Conference statement on Bariatric Surgery for Morbid Obesity which discussed the expansion of available operative procedures, the growth in laparoscopic techniques, and the significant reductions in perioperative morbidity and mortality that have occurred since the 1991 NIH statement. In the years following the 2004 ASBS statement, societies including the American Society for Metabolic and Bariatric Surgery (ASMBS), the Obesity Society, the American Association of Clinical Endocrinologists (AACE), and the International Diabetes Federation (IDF) have endorsed consideration of lower BMI patients (30–34.9) for metabolic and bariatric operations [18]. Surgical Technique The introduction of one anastomosis gastric bypass into our practice evolved from our extensive experience with Roux-Y gastric bypass, handsewn and stapled anastomosis, and revisional procedures. Experience with laparoscopic anastomosis is essential when performing OAGB. Complications that can develop from a poorly done anastomosis can result in intractable reflux, outlet obstruction, afferent loop syndrome, and bile leakage. Surgeons whose experience is limited with respect to laparoscopic suturing of gastrojejunal anastomosis are encouraged to spend time improving their skills with respect to anastomotic technique. Advanced laparoscopic suturing skills are required to manage complications of these procedures. Surgeons with limited experience in performing laparoscopic suturing will find themselves at a disadvantage when performing the OAGB procedure or managing complications that may occur at the gastrojejunostomy. Educational courses featuring experts and faculty with extensive experience performing OAGB are becoming more widely available. All patients receive a form of subcutaneous heparin and pneumatic compression devices to prevent DVT formation during the surgery. Patient positioning can either be via the French or split-leg positioning or standard supine position. We have performed our procedures using both techniques and find that either positioning is adequate, and there is relatively no advantage offered by one positioning technique H. T. Billy et al. over the other. Arm positioning is at 90° to the operating table, and padded straps to secure the lower extremities are used in all procedures as well as a foot rest to prevent patient movement and sliding during the procedure. Access to the peritoneum is performed using an optical 12 mm trocar technique at the umbilicus utilizing a paraumbilical crease for the access incision. Establishment of pneumoperitoneum is followed by placement of a right upper quadrant, subcostal 12 mm trocar. Two additional trocars are positioned slightly higher than utilized in traditional RYGB operations and include a 5 mm port in the left upper lateral abdomen and a 12 mm port in the left mid-abdomen lateral to the umbilicus. We prefer a 10 mm 30° operating laparoscope although surgeons preferring a 5 mm scope could utilize that and downsize the umbilical port to a 5 mm as per their preference. If the patient is positioned supine, the surgeon is standing on the patient’s right side. The majority of our patients have been placed on a 2-week liquid diet to facilitate reduction in the size of the liver. In most cases, the decrease in liver size will be sufficient so as to allow the use of internal liver retraction devices which typically suspend the liver using three hooks attached to suture technique. A deceased liver size avoids the deployment of external retraction devices such as the Nathanson hook which requires an additional incision and significant torque on the liver in order to provide exposure. We have also subjectively observed less congestion of the liver and less aggressive retraction using internal liver retraction. Creating the Gastric Pouch Creation of the gastric pouch begins with a lesser curve dissection and identification of the crow’s foot as the critical landmark to insure optimal pouch length in order to minimize any likelihood that refluxed biliopancreatic secretions could reach the esophagus (Fig. 16.3). The limitations of the Mason loop gastric bypass were largely due to the combination of a short pouch which was horizontal in nature with a loop reconstruction [19]. In order to perform a successful OAGB, and minimize any biliopancreatic reflux, the identification of the crow’s foot of the lesser curve is the key to a successful operation. The majority of surgeons begin their perigastric dissection at the crow’s foot although we prefer beginning our dissection 1–2 cm or so below the crow’s foot (Fig. 16.4). Our preferred bougie is a 34 French oral gastric tube, but we have used 36 and even 40 French bougies when necessary. Unlike a sleeve gastrectomy, the goal when creating the gastric pouch is to avoid hugging the bougie and to create a pouch in which the stapler is positioned a bit wider and lateral to the bougie. Once the perigastric dissection has been completed and the lesser sack has been entered, we position a 45 mm linear 16 Laparoscopic One Anastomosis Gastric Bypass: History of the Procedure Surgical Technique and Outcomes Fig. 16.3 Operative inspection of the stomach. Key landmarks include the crow’s foot and the planned path for stapling to create the gastric pouch as well as the pylorus Fig. 16.4 Dissection for the initial path of the stapler occurs 1–2 cm below the crow’s foot along the lesser curve stapler via the left upper quadrant 12 mm port just below the crow’s foot (1–2 cm) and perpendicular to the antrum (Fig. 16.5). The initial firing is with a cartridge used for the thickest tissue. Caution is taken so as to not transect the antrum or narrow the outflow of what will be the retained body and fundus of the bypassed portion of the stomach. A 60 cm linear stapler is then introduced through the left upper quadrant 12 mm port and positioned so as to create a staple line curving toward the angle of His while staying a bit lateral to the 34 French bougie and not hugging the bougie as is typically done with sleeve gastrectomy (Fig. 16.6). The initial firing is again with a cartridge used for the thickest tissue, and then adjustments in cartridge selection can occur based on the thickness of the tissue encountered. We consider bioabsorbable buttressing to limit staple line bleeding 185 Fig. 16.5 Initial positioning of the first staple cartridge 1–2 cm below the crow’s foot for one anastomosis gastric bypass Fig. 16.6 Positioning of the second staple firing for creation of the gastric pouch for one anastomosis gastric bypass on all firings after the first 60 mm firing. Multiple 60 mm cartridges are deployed until the stomach is transected just lateral to the angle of His separating the long gastric pouch from the gastric remnant. It is essential to stay a bit wide of the bougie in order to insure that staple cartridges are positioned in the same horizontal plane and at the apex of each staple firing. This careful attention to detail is to prevent a spiral of the staple line which can result in a functional obstruction and severe reflux. We do not routinely oversew our staple lines and address any persistent bleeding with additional sutures or surgical clips. Creation of the loop begins by revealing and exposing the ligament of Treitz. We use a 200 cm afferent limb for patients with a BMI under 50 and consider 250 cm afferent limb for patients with a BMI over 50. We utilize bowel graspers with 186 Fig. 16.7 Completion of the initial suture line in preparation of a side-­ to-­side linear stapled anastomosis for one anastomosis gastric bypass marks signifying 5 cm and 10 cm lengths to measure as precisely as possible, and we do not estimate the measurements via visual estimations. Once a 200 cm afferent limb has been measured, we favor the anti-reflux technique described by Cabajo and Caballero utilizing a side-to-side anastomosis of the afferent limb to the distal pouch. The gastric pouch which is typically 15–18 cm in length is positioned adjacent to the afferent limb to create a 200 cm biliopancreatic limb length. The site of the 200 cm measurement is marked with a stay suture, and measurements are continued distally to insure that there is at least 250–300 cm of small bowel distal to the proposed site of the gastrojejunostomy. Once the site of the 200 cm afferent loop is confirmed, a posterior row of suture using 3–0 absorbable suture is placed to approximate the mesenteric serosa of the afferent limb to the lateral suture line of the distal pouch (Fig. 16.7). This suture line not only protects from leaks but also is essential in taking any unnecessary tension off the staple line. Once complete, an opening is made in the distal gastric pouch and the antimesenteric portion of the afferent limb so as to allow positioning of a 45 mm stapler to create a 3–4 cm gastrojejunal anastomosis. The stapler is positioned intraluminally in the gastric pouch and the afferent loop and then fired (Fig. 16.8). The 34 French bougie is advanced across the anastomosis and the remaining gastrojejunal defect is closed anteriorly with 3–0 absorbable suture followed by a second layer of 3–0 absorbable suture to oversew the anastomosis and approximate the serosa of the afferent limb to the serosa of the gastric pouch (Fig. 16.9). By utilizing this technique, the initial running suture line suspends the afferent limb above the anastomosis and secures the loop to the gastric pouch. We believe this technique not only will decrease the incidence of symptomatic bile reflux but will also decrease the possibility of developing an afferent loop syndrome postoperatively. H. T. Billy et al. Fig. 16.8 Intraluminal positioning of linear stapler for a side-to-side 2.5–4 cm stapled anastomosis one anastomosis gastric bypass Fig. 16.9 Handsewn closure of stapled side-to-side anastomosis, one anastomosis gastric bypass The anastomosis and vertical staple line are tested for leaks by occluding both afferent and efferent limb and insufflating with oxygen or air under saline submergence. Any evidence of leak or visible air bubbles are addressed using intracorporeal suturing until there is no evidence of any air leak. Caution must be taken not to over-distend the pouch and afferent limb, and rarely do we need to insufflate with a pressure greater than 2 L per minute inflow. There must be excellent communication between the surgeon and the anesthesiologist during the leak test to avoid any rupture of the bowel or anastomosis due to over-distension. Alternatively, we have utilized the injection of methylene blue diluted into 1 L of saline as an alternative. Injection of 100–150 cc of diluted methylene blue is done by our anesthesiologist to accomplish distension of the gastric pouch as well as a short segment of the afferent and efferent limbs. Both limbs are 16 Laparoscopic One Anastomosis Gastric Bypass: History of the Procedure Surgical Technique and Outcomes 187 5. Measure 200 cm distal to the ligament of Treitz, and anastomose the loop to the gastric pouch in a side-to-side fashion with the afferent limb superior and the efferent limb inferior. 6. Close the mesenteric defect. Postoperative management is similar to traditional gastric bypass. We keep our patients overnight and start them immediately on a clear liquid diet. Almost all patients can be discharged postoperative day 1. We do not use routine upper GI swallows immediately post-op. Reversal of One Anastomosis Gastric Bypass Fig. 16.10 Same patient 1 year after one anastomosis gastric bypass. Visualized during laparoscopic cholecystectomy. The gastrojejunostomy is easily visualized below the liver and sits close to the transverse colon. The antrum and previous staple line are also visualized. EGD revealed no evidence of bile reflux, ulcer, or stricture. One year post-op, this patient has achieved a BMI of 25 and has no complaints occluded by laparoscopic bowel clamps during the air insufflation or methylene blue tests. Finally, we choose to close the mesenteric defect created by the afferent limb between the root of the mesentery of the afferent limb and the transverse mesocolon. Because the loop is an antecolic path the creation of a mesenteric defect between the mesentery of the loop and the transverse mesocolon is a recognized site of potential internal hernia. The defect is addressed using 3–0 nonabsorbable suture. The root of the mesentery of the loop is identified and sutured to the mesentery of the transverse colon until the mesenteric defect is closed. After completing a final secondary look for bleeding or leaks, the retractors are removed, and the 12 mm ­fascial defects are closed, and the procedure is completed (Fig. 16.10). Our technique can be summarized by identifying several steps as all our OAGB procedures are done in the same manner: 1. Begin a perigastric dissection 1–2 cm distal to the crow’s foot along the lesser curve. 2. Position a 45 mm stapler using a staple cartridge for thick tissue at the point of the perigastric dissection staying perpendicular to the lesser curve of the stomach. 3. Position a 34 French bougie intragastric followed by a 60 cm stapler just lateral to the bougie. 4. Transect just lateral to the angle of His to create a somewhat generous and straight gastric pouch. Postoperative complications following one anastomosis gastric bypass procedures are a rare but recognized reason for considering reversal of the procedure. Malnutrition, excessive weight loss, severe lower extremity edema, and motor deficits can present as a refractory malnutrition syndrome after any malabsorptive procedure. Genser et al. reported on a series of 2934 patients who had undergone mini gastric bypass over a 10-year period [20]. Of the 2934 patients, 26 were identified as having developed severe and refractory malnutrition syndrome which responded to reversal of MGB to normal anatomy. Even with variations in surgical technique, reversal of a one anastomosis gastric bypass is straightforward allowing for complete restoration of normal anatomy. There are essentially three steps involved in reversing an OAGB operation: 1. Identification of the gastrojejunostomy 2. Resection of the gastrojejunostomy 3. Anastomosis of the gastric pouch to the body of the gastric remnant Identification and dissection of the gastrojejunostomy away from adjacent structures allow for a straightforward transection of the distal gastric pouch disconnecting the stomach from the afferent limb. Following this, the gastrojejunostomy can be similarly removed from the afferent limb by careful placement of a 60 cm stapler along the antimesenteric border of the jejunum and then resecting the anastomosis off of the jejunum. Once the gastrojejunal anastomosis has been resected, all that remains is to reestablish continuity between the gastric pouch and the gastric remnant. We accomplish this by approximating the two stomachs using an absorbable 3–0 suture and then completing the anastomosis with a linear stapler followed by closure of the new anastomosis over a bougie using 3–0 absorbable suture in two layers. The anastomosis is tested for leaks using methylene blue or air insufflation and the reversal is completed. Patients are started on clear liquids postoperatively and are discharged after an overnight stay. 188 H. T. Billy et al. Benefits of a Single Anastomosis Procedure Mesenteric Defects Mesenteric defects created by RYGB are a potential source of internal hernia, bleeding, and bowel obstruction. Obstruction from internal hernia following Roux-Y gastric bypass is relatively common. Internal hernia as the primary cause of obstruction following gastric bypass represents up to 41% of cases presenting with intestinal obstruction [21]. Roux-Y gastric bypass requires the creation of a gastrojejunostomy and a second enteroenterostomy. Depending on whether an antecolic approach or a retrocolic approach is undertaken, there can be up to 3 potential spaces where internal herniation can occur. OAGB eliminates the need for the enteroenterostomy created when a RYGB is performed. The second anastomosis which is created between the biliopancreatic limb and the alimentary limb is unique to RYGB and creates a potential site for internal hernia that does not exist in an OAGB operation. The herniation of bowel through this kind of defect creates a surgical emergency that must be evaluated quickly to avoid bowel ischemia and infarction. Closure of mesenteric defects can reduce the risk of internal hernia from 3.3% to 1.2% but cannot eliminate the risk [22]. If a retrocolic passage of the Roux limb to gain access to the gastric pouch is used, a third potential site of herniation through the mesenteric defect is created which also must be closed. The simplicity of the OAGB eliminated two of these three potential sites of internal herniation. As a result, the potential for internal hernia is lower in OAGB than in RYGB. Internal hernia following OAGB is a rare complication [23, 24]. The benefit of decreasing the potential defects associated with internal hernia following gastric bypass is not new. The modification of the retrocolic gastric bypass to the antecolic technique of passage of the Roux limb from the midgut to the foregut eliminated one of the three potential sites of internal hernia [25, 26]. As a result, the risk of internal hernia through the mesentery of the transverse colon was eliminated, reducing morbidity and mortality from internal herniation [27, 28]. OAGB has the benefit of decreasing morbidity and mortality further by eliminating one of the two remaining sites of potential internal herniation that exist in RYGB. Internal hernia rates following RYGB have been reported. Iannelli et al. reported findings of 11,918 patients following RYGB. Internal herniation was discovered in 300 patients for an internal hernia rate of 2.51% [29]. Internal herniation occurred at the level of the transverse colon in 69% of cases, at the level of the Peterson’s defect in 18% of cases, and at the level of enteroenterostomy in 13% of cases. OAGB creates no defect at the level of the transverse colon or at the enteroenterostomy. Eighty-two percent of the cases of internal hernias reported in RYGB are associated with these two defects. Internal hernia reported with OAGB are relatively rare compared to the incidence reported with RYGB [30]. Fig. 16.11 Closure of the only mesenteric defect in one anastomosis gastric bypass is accomplished by closing the defect between the efferent limb and the mesentery to the transverse colon. Nonabsorbable suture is used to close the defect beginning at the root of the mesentery and approximating it to the transverse mesocolon until the lower border of the colon is reached The OAGB utilizes a single gastrojejunal anastomosis with a single mesenteric Peterson hernia defect created by the loop anastomosis of jejunum to gastric pouch. Closure of this mesenteric defect is relatively easy and is associated with significant decrease in the rate of internal hernia from this single remaining site (Fig. 16.11). Intussusception Jejuno-jejunal intussusception is a known complication following RYGB. Obstruction, bowel ischemia, and bowel necrosis are potential sequela following development of intussusception. The rate of intussusception following RYGB at the level of the enteroenterostomy occurs at less than 1% (0.4%) [31]. Diagnosis can be difficult as symptoms are generally nonspecific. Both operative and nonoperative approaches to the treatment of intussusception do not reliably prevent recurrence of the problem. The single anastomosis OAGB eliminates the potential of intussusception of the enteroenterostomy further decreasing postoperative morbidity and mortality from RYGB-related complications. Simplified Reversal/Revision Reversal of OAGB can be accomplished via two approaches. Complete anatomic reversal requires simple transection of the gastrojejunostomy on the jejunal side of the anastomosis followed by creation of a gastro-gastrostomy between the gastric pouch and the gastric remnant. Alternatively, func- 16 Laparoscopic One Anastomosis Gastric Bypass: History of the Procedure Surgical Technique and Outcomes tional reversal of the OAGB can be accomplished by simply reconnecting the stomach to the gastric remnant. Conversion to Roux-Y Configuration In the rare event that bile reflux is suspected, diversion of the bile away from the gastrojejunostomy can be accomplished by simply transecting the afferent limb at the level of the gastrojejunostomy and diverting the flow at least 50–70 cm below the gastrojejunostomy utilizing a new enteroenterostomy. Complications Bile Reflux Perhaps the most contentious, widely discussed topic surrounding one anastomosis gastric bypass has been the issue of potential bile reflux (Figs. 16.12 and 16.13). The topic remains controversial with potential concerns regarding Barrett’s esophagus and possible esophageal cancer persisting although no reported cases of esophageal cancer appear in peer reviewed publications. Despite the large number of publications and low incidence of reflux-related revisions, this controversy continues to persist and has resulted in a 189 delay of acceptance of the procedure particularly in the United States. Most authors report an improvement in GERD symptoms following OAGB [32]; however, some authors report reflux symptoms that have required revision to either RYGB or implementation of Braun’s anastomosis distal to the gastrojejunostomy of the single anastomosis of the gastrojejunostomy [33]. The incidence of symptomatic acid reflux in our patients that have undergone OAGB procedures is higher than what we have experienced in our RYGB patients. The majority of these patients are easily treated with once-a-day PPI therapy, and many reflux symptoms dissipate over time. The few patients that have had significant reflux symptoms following OAGB have been successfully revised to a Roux-Y gastric bypass with complete resolution of their symptoms. Many surgeons have reported an incidence of symptomatic reflux following one anastomosis gastric bypass at less than 0.5%. Despite positive effect in terms of weight loss and improvement of obesity-related comorbidities, there still exist concerns about symptomatic biliary reflux gastritis and esophagitis requiring revision surgery following OAGB procedures [34]. Concerns regarding the risk of esophageal cancer with this procedure because of chronic biliary reflux persist [35]. There is no prospective data to suggest there is a legitimate concern for gastroesophageal reflux following OAGB to progress to Barrett’s esophagus or cancer. Fig. 16.12 Classic bile reflux following mini gastric bypass performed as described by Rutledge in a patient with previous repair of a large para esophageal hiatal hernia and poorly functioning lower esophageal sphincter 190 H. T. Billy et al. complaints of reflux occurring in patients having had no history of hiatal hernia or reflux symptoms prior to surgery. Our only two findings of significant reflux have occurred in two patients both of whom had significant abnormalities of the distal esophagus and esophagogastric junction prior to their OAGB. It has been our experience that patients with symptomatic bile reflux can be confirmed on endoscopic examination and can be successfully treated with revision to a Roux-Y configuration if necessary with complete resolution of symptoms [37]. Bile Scintigraphy Fig. 16.13 Esophageal view, bile reflux following mini gastric bypass. Same patient as Fig. 16.12 Tolone et al. evaluated 15 patients (5 males/10 females), with a mean age of 38 years, a median preoperative weight of 141.1 kg (121–174), and a mean BMI of 46.4 (38–60) kg/m2, who had undergone OAGB [36]. At the 1-year postoperative follow-up, the median weight was 81.2 kg (72–111), the median BMI was 31 kg/m2 (28–42), and the excess weight loss (EWL, %) was 63 (56–69). The OAGB was compared to a control group consisting of 25 adult patients who underwent sleeve gastrectomy. The sleeve gastrectomy group was age- and sex-matched with patients who underwent OAGB. Their median preoperative weight was 130.8 kg (119–156), and median BMI was 46.1 (38–58); 1-year postoperative median weight was 98 kg (72–110), and median BMI was 34.7 (28–46), with 56% excess weight loss. In this study, the OAGB group (15 patients) reported no symptoms of reflux prior to surgery. Following OAGB, none of the 15 patients reported reflux/GERD symptoms, and there was no esophagitis on endoscopic examination. Using high-resolution impedance manometry and 24-h pHimpedance monitoring, Tolone discovered a significant reduction in esophageal acid exposure as well as reflux episodes in all patients undergoing OAGB. The control group of patients undergoing sleeve gastrectomy developed an increase in both esophageal acid exposure and reflux episodes. They concluded that patients undergoing OAGB developed no changes in esophageal gastric junction function or motility patterns in obese patients without preoperative GERD or large hiatal hernia 12 months after surgery. In contrast to patients having undergone sleeve gastrectomy, those who had undergone OAGB demonstrated diminished esophageal acid exposure and total number of reflux. Although this study reports a rather small patient population, it remains perhaps the most elegant and complete study exploring the function of the esophageal gastric junction following OAGB operations. Our experience with bile reflux following OAGB has been similar to many published studies with virtually no Bile scintigraphy has been published and is a possible modality to assess the presence of bile reflux in patients following mini gastric bypass [38] (Fig. 16.14). In this study, nine consecutive patients who underwent OAGB underwent hepatobiliary scintigraphy and a reflux questionnaire 8–13 months after their operations. In addition, every patient with a positive bile scintigraphy scan underwent gastroscopy with biopsies. Five of the nine patients demonstrated positive reflux into the gastric tube; however, bile reflux was not present in the esophagus of any of the nine patients. The findings demonstrated that transient bile reflux was common after MGB but only in the gastric tube and not in the esophagus. The clinical relevance of bile reflux and loop bariatric procedures has not been adequately studied. The debate regarding OABG procedures and the clinical significance of reflux following these procedures will continue, and further prospective studies both short-term and long-term are necessary to further define the clinical relevance of bile reflux. Malabsorption Multiple studies have confirmed that malabsorption can occur following OAGB, and careful postoperative monitoring of nutritional status is recommended following this procedure [39]. Unlike Roux-Y gastric bypass where no standardization of the alimentary, biliopancreatic, and common limb lengths has been defined, OAGB procedures typically describe a biliopancreatic alimentary limb length of between 150 and 200 cm. Nutritional outcomes utilizing these limb lengths have been extensively reported [40, 41]. One hundred fifty centimeters of limb length has been proven to be effective in patients with class II obesity. Regardless of the method used to determine if the limb length should be 150 cm or 200 cm, a risk of revisional surgery for excess weight loss was reported by Noun to be 0.4% [42]. Lee reported a 1.0% and Musella et al. [43] reported an incidence of excess weight loss at 0.2%. Authors who have used a fixed 200 cm biliopancreatic loop in the creation of a one anasto- 16 Laparoscopic One Anastomosis Gastric Bypass: History of the Procedure Surgical Technique and Outcomes 191 Fig. 16.14 Nuclear medicine bile scintigraphy following one anastomosis gastric bypass demonstrates no significant bile reflux. The scan correlates with the endoscopic findings which were devoid of any bile reflux in the gastric pouch or esophagus. Arrow depicts very small uptake in the gastric pouch but below the diaphragm which may represent trace amounts of bile reflux but no significant reflux entering the distal esophagus mosis gastric bypass report a much lower rate of malnutrition of 0.05% and 0.1% by Chevallier and Kular [44]. ditions following OAGB/MGB was published in 2019. In it, type 2 diabetes mellitus remission rate was 83% and 70% at 1 and 3 years, respectively. Hypertension resolution was 61%, 58%, and 58% at 1, 2, and 3 years post-op. Ninety-nine percent of sleep apnea patients improved symptomatically and went off their CPAP machines. The study reported on outcomes of 527 OAGB patients. Mean follow-up was 2.5 years and the mortality was zero. Multiple additional authors report similar results with comorbidity resolution following laparoscopic OAGB and similar procedures [42, 45–50]. esolution of Comorbidities and Nutritional R Complications The major comorbid conditions commonly associated with one anastomosis gastric bypass which include diabetes, hypertension, obstructive sleep apnea, hyperlipidemia, and joint pain all show significant improvement after 5 years of follow-up. Bruzzi et al. published a series of 126 patients with 72% follow-up at 5 years [46]. No mortalities were reported, and percent excess body mass index lost was an impressive 71%. Complete remission of type 2 diabetes occurred in 82% of patients. Fifty-two percent of patients with hypertension and 81% of patients with hyperlipidemia were able to stop medications at 5 years follow-up. Sleep apnea resolved in 50% of patients. The largest UK study showing safety and acceptable results for metabolic syndrome and obesity comorbid con- utcome Comparison Roux-Y Gastric Bypass O vs Sleeve Gastrectomy vs One Anastomosis Gastric Bypass The most recent prospective randomized series comparing long-term results between Roux-Y gastric bypass versus sleeve gastrectomy versus one anastomosis gastric bypass was recently published by Ruiz-Tovar et al. [51]. In this 192 study, 600 patients undergoing primary bariatric surgery were prospectively randomized into a clinical trial consisting of three groups. Patients were randomized into those undergoing sleeve gastrectomy (SG), Roux-Y gastric bypass (RYGB), and one anastomosis gastric bypass (OAGB). The study reports on the long-term findings with respect to BMI, excess BMI loss, remission of type 2 diabetes, hypertension, and dyslipidemia. Six hundred patients were randomized into three groups, 200 patients in each group. The authors achieved 91% follow-­up at 5 years in the SG group, 90% follow-up in the OAGB group, and 92% follow-up in the RYGB group. The study reports significantly greater excess BMI loss in the OAGB group versus that achieved in the SG and RYGB group at 5 years follow-up. Similarly the OAGB group achieved greater remission of type 2 diabetes, hypertension, and dyslipidemia than RYGB or SG. The results build upon findings observed with previous studies and continue to confirm the findings that OAGB is a safe and efficacious operation. Prospective randomized studies continue to confirm short-term and long-term outcomes that meet or exceed those of SG and RYGB. The importance of these studies in contributing to the worldwide adoption of the OAGB as an acceptable, safe, and effective primary bariatric operation cannot be underestimated. Insurance Coverage USA Insurance coverage for one anastomosis gastric bypass procedures has been slow to obtain adoption by commercial insurance companies in the United States. At the current time, there are no know commercial payers in the United States who have included MGB, OAGB, or single anastomosis gastric bypass as a covered benefit for bariatric surgery. In our review of over 20 different benefits of coverage policies for virtually every major insurance company in the United States, we were unable to find a single policy which did not specifically exclude these procedures, and in every instance they were identified as being investigational. In every policy reviewed, the OAGB procedures, procedures using a loop gastrojejunostomy or Billroth II-type anastomosis, or procedures described as mini gastric bypass, for the purpose of weight loss, were specifically identified as procedures which were not a covered benefit of the insurance plan with respect to the surgical treatment of morbid obesity. Summary One anastomosis gastric bypass/mini gastric bypass is clearly a promising bariatric operation, one that still continues to polarize bariatric surgeons. Despite the many thou- H. T. Billy et al. sands of procedures published in the world literature, there remain objections to the one anastomosis procedures. These objections have led to a slow, almost insignificant adoption of the procedure in the United States as the concern regarding symptomatic bile reflux, malnutrition, and risk of esophageal cancer remains the most common objections voiced by those surgeons reluctant to perform this operation. Those surgeons having demonstrated significant expertise with the one anastomosis approach to gastric bypass report favorable outcomes with excellent resolution of comorbid conditions and low complication rates. In addition, the technical simplicity of the operation and short learning curve, standardized approach, and relative ease with which the operation can be reversed or revised lead many surgeons to advocate for a more widespread proliferation of this operation as a preferred operation for the treatment of obesity. Multiple publications including prospective and randomized trials have demonstrated that OAGB/mini gastric bypass is at least comparable to Roux-Y gastric bypass in the treatment of obesity. Our personal experience with OAGB exceeds 5 years, and thus far we have experienced results similar to that described in the world’s body of published literature. The single anastomosis approach to gastric bypass can be performed in less time than a Roux-Y gastric bypass, utilizes less resources, and has a very low complication rate. Time will tell over the next few years if this procedure develops more widespread adoption among American surgeons and if the major insurance plans in the United States will begin to recognize this approach as an effective alternative. Question Section 1. One anastomosis gastric bypass A. Consists of a short gastric tube similar to a RYGB. B. Consists of a long gastric tube created below the crow’s foot of the lesser curve. C. Must consist of a tight narrow gastric tube to avoid weight regain. D. A 40 French Bougie is to be avoided. 2. Malnutrition associated with one anastomosis gastric bypass A. Increases as the afferent biliopancreatic limb exceeds 250–300 cm B. Can be avoided by insuring that there are at least 250– 300 cm of bowel distal to the loop gastrojejunostomy C. Can be improved by revision of the biliopancreatic loop to a 150–200 cm length D. All of the above 3. Reversal of one anastomosis gastric bypass A. Requires small bowel resection and reconstruction into a Roux-Y configuration 16 Laparoscopic One Anastomosis Gastric Bypass: History of the Procedure Surgical Technique and Outcomes 193 (OAGB/MGB) using a modified Delphi approach. Obes Surg. 2018;28(2):303–12. 16. Lee WJ, Lin YH. Single-anastomosis gastric bypass (SAGB): appraisal of clinical evidence. Obes Surg. 2014;24(10):1749–56. 17. Gastrointestinal Surgery for Severe Obesity. Consens Statement. 1991;9(1):1–20. 18. Shauer P, Rubino F. International diabetes federation position statement on bariatric surgery for type 2 diabetes: implications for patients, physicians and surgeons. Surg Obes Relat Dis. 2011;7(4):448–51. 19. Mason E, Printen K, Lewis J, et al. Gastric bypass criteria for effectiveness. Int J Obes. 1981;5(4):405–11. 20. Genser L, Soprani A, Tabbara M, et al. Laparoscopic reversal of mini gastric bypass to original anatomy for severe postoperative malnutrition. Langenbeck’s Arch Surg. 2017;402(8):1263–70. 21. Khoraki J, Mazzini G. Early small bowel obstruction after laparoscopic gastric bypass: a surgical emergency. Surg Obes Relat Dis. 2018;14(8):1118–25. 22. Blockhuys M, Gypen B, Heyman S, et al. Internal hernia after laparoscopic gastric bypass: effect of closure of the Petersen defect – single-center study. Obes Surg. 2019;29(1):70–5. 23. Kermansaravi M, Kazazi M, Pazouki A. Petersen’s space internal References hernia after laparoscopic one anastomosis (mini) gastric bypass. Cas Rep Surg. 2018;2018:9576120. 1. Rutledge R. The mini-gastric bypass: experience with the first 24. Genser L, Carandina S, Soprani A. Petersens’s internal hernia 1,274 cases. Obes Surg. 2001;11(3):276–80. complicating a laparoscopic omega loop gastric bypass. Surg Obes 2. Mason E. Gastric bypass in obesity. 1967. Obes Res. Relat Dis. 2015;11(3):e33–4. 1996;4(3):301–3. 25. Garza E, Kuhn J, Arnold D. Internal hernias after laparoscopic 3. DeMeester S, DeMeester T. Barrett’s esophagus. Current Prob Roux-en-Y gastric bypass. Am J Surg. 2004;186(6):796–800. Surg. 2001;38:549–640. 26. Higa K, Ho T, Boone K. Internal hernias after laparoscopic Roux-­ 4. Theisen J, Peters J, Fein M, Hughes M, Hagen J, DeMeester T, en-­Y gastric bypass: incidence, treatment and prevention. Obes Laird P. The mutagenic potential of Duodenoesophageal reflux. Surg. 2003;13(3):350–4. Ann Surg. 2005;241(1):63–8. 27. Al-Mansour M, Mundy R, Canoy J, et al. Internal hernia after 5. Genco A, Soricelli E, Casella G, et al. Gastroesophageal reflux dislaparoscopic antecolic Roux-en-Y gastric bypass. Obes Surg. ease and Barrett’s esophagus after laparoscopic sleeve gastrectomy: 2015;25(11):2106–11. a possible, underestimated long-term complication. Surg Obes 28. Kristensen S, Jess P, Floyd A, et al. Internal herniation after lapaRelat Dis. 2017;13(4):568–74. roscopic antecolic roux-en-Y gastric bypass: a nationwide Danish 6. Parmar C, Mahawer K. One anastomosis gastric bypass is now study based on the Danish National Patient Register. Surg Obes an established bariatric procedure: a systemic review of 12,807 Relat Dis. 2016;12(2):297–303. patients. Obes Surg. 2018;28(9):2956–296. 29. Iannelli A, Facchiano E, Gugenheim J. Internal hernia after lapa7. Higa K, Brown W, Himpens J. Editorial: single anastomosis proceroscopic Roux-en-Y gastric bypass for morbid obesity. Obes Surg. dures, IFSO position statement. Obes Surg. 2018;28(5):1186–7. 2006;16(10):1265–71. 8. De Luca M, Tie T, Ooi G, Higa K, Himpens J, Cabajo M, Mahawar 30. Kular K, Prasad A, Ramana B. Petersen’s hernia after mini (one K, Shikora S, Brown W. Mini Gastric Bypass-One Anastomosis anastomosis) gastric bypass. J Visc Surg. 2016;153(4):321. Gastric Bypass (MGB-OAGB)-IFSO position statement. Obes 31. Edwards M, Grinbaum R, Ellsmere J. Intussusception after Roux-­ Surg. 2018;28(5):1188–206. en-­Y gastric bypass for morbid obesity: case report and literature 9. Mukherjee M, Devalia M, Mehtab G. Sleeve gastrectomy as a review of rare complication. Surg Obes Relat Dis. 2006;2(4):483–9. bridge to a second bariatric procedure in superobese patients-a sin- 32. Wang F, Yan W, Song M. Outcomes of mini vs Roux-en-Y gasgle institution experience. Surg Obes Relat Dis. 2012;8(2):140–4. tric bypass: a meta-analysis and systemic review. Int J Surg. 10. Madhok B, Mahawar KK, Boyle M, et al. Management of 2018;56:7–14. super-super obese patients: comparison between mini (one anas- 33. Johnson W, Fernanadez A, Farrell T, et al. Surgical revision of loop tomosis) gastric bypass and sleeve gastrectomy. Obes Surg. (“mini”) gastric bypass procedure: multicenter review of compli2016;26(7):1646–9. cations and conversions to Roux-en-Y gastric bypass. SOARD. 11. Navarrette S, Leyba J, Li S, et al. Results of the comparative study 2007;3:37–41. of 200 cases: one anastomosis gastric bypass vs Roux-en-Y gastric 34. Bruzi M, Chevallier J, Czernichow S. One-anastomosis gasbypass. Obes Surg. 2018;28(9):2597–602. tric bypass: why biliary reflux remains controversial. Obes Surg. 12. Rutledge R, Kular K, Manchanda N. The mini-gastric bypass origi2017;27(2):545–7. nal technique. Int J Surg. 2019;61:38–41. 35. Mahawar K, Borg C, Kular K. Understanding objections to one 13. Ahuja A, Tantia O, Goyal G, et al. MGB-OAGB: effect of bilioanastomosis (mini) gastric bypass: a survey of 417 surgeons not pancreatic limb length on nutritional deficiency, weight loss and performing this procedure. Obes Surg. 2017;27(9):2222–8. comorbidity resolution. Obes Surg. 2018;28(11):3439–45. 36. Tolone S, Cristiano S, Savararino E, et al. Effects of omega-loop 14. Carbajo M, Garcia-Caballero M, Toledano M, et al. One-­ bypass on esophagogastric junction function. Surg Obes Relat Dis. anastomosis gastric bypass by laparoscopy: results of 209 patients. 2016;12(1):62–9. Obes Surg. 2005;15(3):398–404. 37. Armor I, Petrucciani N. Kassir, et al. laparoscopic conversion of 15. Mahawar K, Himpens J, Shikora S, Chevallier J, et al. The first one anastomosis gastric bypass to a standard Roux-en-Y gastric consensus statement on One Anastomosis/Mini Gastric Bypass bypass. Obes Surg. 2017;27(5):1398. B. Can be accomplished without resection of the gastrojejunostomy C. Leads to intractable bile reflux D. Discovery of Barret’s esophagus postoperatively is the primary indication for reversal 4. Bile reflux following one anastomosis gastric bypass A. Is a common complication leading to Barrett’s esophagus and early esophageal cancer B. Can be prevented by the administration of double dose PPI therapy C. Can be eliminated by revision to a Roux-Y configuration D. Is difficult to detect without annual endoscopy and distal esophageal biopsies 194 38. Saarinen T, Rasanen J, Salo J, et al. Bile reflux scintigraphy after mini-gastric bypass. Obes Surg. 2017;27(8):2083–9. 39. Chevallier JM, Arman GA, Guenzi M, et al. One thousand single anastomosis (omega loop) gastric bypasses to treat morbid obesity in a 7-year period: outcomes show few complications and good efficacy. Obes Surg. 2015;25(6):951–8. 40. Lee WJ, Yu PJ, Wang W, et al. Laparoscopic roux-en-Y versus mini-­ gastric bypass for the treatment of morbid obesity: a prospective randomized controlled clinical trial. Ann Surg. 2005;242(1):20–8. 41. Lee WJ, Ser KH, Lee YC, et al. Laparoscopic roux-en-Y vs. mini gastric bypass for the treatment of morbid obesity: a 10-year experience. Obes Surg. 2012;22(12):1827–34. 42. Noun R, Skaff J, Riachi E, et al. One thousand consecutive mini gastric bypass short and long-term outcomes. Obes Surg. 2012;22:697–703. 43. Musella M, Milone M. Still “controversies” about the mini gastric bypass? Obes Surg. 2014;24(4):643–4. 44. Kular KS, Manchanda N, Rutledge R. A 6-year experience with 1,054 mini-gastric bypasses-first study from an Indian subcontinent. Obes Surg. 2014;24(9):1430–5. 45. Hussain A, El-Hasani S. Short and mid-term outcomes of 527 One Anastomosis Gastric Bypass/Mini-Gastric Bypass (OAGB/MGB) operations: retrospective study. Obes Surg. 2019;29(1):262–7. H. T. Billy et al. 46. Bruzzi M, Rau C, Voron T, Guenzi M, Berger A, Chevallier JM. Single anastomosis or mini gastric bypass; long-term results and quality of life after a 5 year follow-up. Surg Obes Relat Dis. 2015;11(2):321–6. 47. Lee WJ, Ser KH, Lee YC, Tsou JJ, Chen SC, Chen JC. Laparoscopic Roux-en Y vs mini gastric bypass for the treatment of morbid obesity, a 10 year experience. Obes Surg. 2012;22:1827–34. 48. Rutledge R, Walsh TR. Continued excellent results with the mini gastric bypass: six-year study in 2410 patients. Obes Surg. 2005;15:398–404. 49. Muscella M, Susa A, Greco F, et al. The laparoscopic mini-gastric bypass: the Italian experience: outcomes from 974 consecutive cases in a multicenter review. Surg Endosc. 2014;28:156–63. 50. Garcia-Caballero M, Valle M, Martínez-Moreno JM, Miralles F, Toval JA, Mata JM, Osorio D, Mínguez A. Resolution of diabetes mellitus and metabolic syndrome in normal weight 24–29 BMI patients with one anastomosis gastric bypass. Nutr Hosp. 2012;27(2):623–31. 51. Ruiz-Tovar J, Carbajo MA, Castro MJ, et al. Long-term follow-up after sleeve gastrectomy versus Roux-en-Y gastric bypass versus one-anastomosis gastric bypass: a prospective randomized comparative study of weight loss and remission of comorbidities. Surg Endosc. 2019;33(2):401–10. Part III Management of Bariatric Complications Management of Gastrointestinal Leaks and Fistula 17 Ninh T. Nguyen and Shaun C. Daly Chapter Objectives 1. Recognize signs and symptoms of gastrointestinal leaks after bariatric surgery. 2. Understand the management of acute gastrointestinal leaks after bariatric surgery. 3. Understand the management of chronic gastrointestinal fistula after bariatric surgery. Introduction Bariatric operations involving gastrointestinal resection and/ or reconstruction can be associated with gastrointestinal leaks and fistula. Common bariatric operations that can lead to development of gastrointestinal leaks include Roux-en-Y gastric bypass, sleeve gastrectomy, and the duodenal switch operation. Of these procedures, sleeve gastrectomy is the most commonly performed operation in the USA. Sleeve gastrectomy as a primary bariatric operation has surpassed Roux-en-Y gastric bypass for the most commonly performed bariatric operation, accounting for 58.2% of cases [1]. The duodenal switch is only being performed at specialized centers and consists of less than 1% of all bariatric procedures performed in the USA. Gastrointestinal leak is one of the most dreaded complications following bariatric surgery as it can lead to significant morbidity and mortality. The incidence of leaks after bariatric surgery varies widely. In a review of the published literature, the mean incidence of leaks after Roux-en-Y gastric bypass was reported to be 0.8%, and the incidence of leaks after sleeve gastrectomy has been reported at 0.7% [2]. N. T. Nguyen · S. C. Daly (*) Department of Surgery, Division of Gastrointestinal Surgery, University of California, Irvine Medical Center, Orange, CA, USA e-mail: shaund@uci.edu Expeditious recognition and early institution of management for gastrointestinal leaks are keys to minimize the progression from systemic inflammatory response to sepsis and septic shock. Recognition of leaks can be particularly difficult in the morbidly obese. A high index of suspicion is crucial in order to recognize these leaks early on. Prompt management of gastrointestinal leak can also minimize the risk for development of a chronic fistula, which is often difficult to treat. This chapter reviews the etiologies of gastrointestinal leaks, common presenting signs and symptoms, diagnostic evaluation, and management of acute gastrointestinal leaks and chronic fistula. Etiologies The causes for leaks are multifactorial and can be divided into technique-related and patient-related factors. Technical factors that can lead to development of leaks include issues such as poor technique in construction of the anastomosis, the presence of excessive tension on the anastomosis, the presence of staple line bleeding, and the presence of tissue ischemia. In most instances, all of the aforementioned factors may play a role in the development of leaks. Patient-­ related factors contributing to the development of leaks include the presence of poor nutrition, current or recent smoking history, liver cirrhosis, and renal failure. Sites for leaks after gastric bypass include the gastrojejunostomy, the gastric remnant, and the jejunojejunostomy with the gastrojejunal anastomosis being the most common site for leaks. The primary reason for the higher leak rate observed at the gastrojejunal anastomosis is the presence of tension at this anastomosis. Additionally, tissue ischemia plays a role both due to the division of the jejunal mesentery, which can compromise tissue perfusion to the antimesenteric jejunum, and the creation of an anastomosis adjacent to a staple line. The site of leak after sleeve gastrectomy can be anywhere along the long gastric staple line, but the most common site of leak, 75–85% of cases, is along the proximal staple line near the © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_17 197 198 N. T. Nguyen and S. C. Daly gastric angle of His. The main reason for leaks in this region is the high intraluminal pressure associated with construction of the sleeve gastrectomy [3]. This phenomenon can be explained by Bernoulli’s principle of fluid dynamics, which states that as fluid moves within a column from an area of higher to lower surface area, the pressure within that column increases. The increased intraluminal pressure can lead to increase in luminal radial force, which can potentially lead to disruption of the staple line. Additionally, the presence of a partial obstruction at the level of the incisura angularis can also lead to additional proximal pressure and can be a contributing factor in the development of leaks after sleeve ­gastrectomy. Other factors that contribute to proximal leaks are the relative tissue ischemia due to ligation of the short gastric vessels and the relative thin tissue of the gastric cardia compared to antral thickness. cumstances. With regard to the vital signs, tachycardia is often the first vital sign to be abnormal [4]. Tachycardia in the early postoperative period should raise suspicion for possible gastrointestinal leaks. Other clinical symptoms include epigastric abdominal pain and fever. Laboratory examination suggestive of a leak includes leukocytosis and an elevated C-reactive protein level. In a study of 17 patients with leaks after gastric bypass, C-reactive protein level of greater than 229 mg/l was able to reliably indicate a leak [5]. CT scan is the radiographic modality of choice for detection of leaks as it can delineate the site of leak and the presence of local or distant fluid/abscess collections (Fig. 17.1). An upper GI study can also be performed for detection of leaks, but unlike CT scan, it will not be able to identify the presence of abdominal fluid collections. CT scan should be performed with PO and IV contrast, as leaks from the gastrojejunal anastomosis or from the sleeve gastrectomy staple line can often be identified by the presence of contrast extravasation. In 2015, the American Society for Metabolic and Bariatric Surgery (ASMBS) published a position statement on prevention and detection of gastrointestinal leak after gastric bypass including the role of imaging and surgical exploration [2]. The sensitivity of upper GI contrast study varies among reports between 22% and 75%. When upper GI and CT are combined, up to one-third of patients with leaks will have both studies interpreted as normal. Therefore, the position statement suggested that laparoscopic or open reexploration should be an appropriate diagnostic option when gastrointes- Presentation and Diagnostic Evaluation Evaluation for gastrointestinal leaks in the postoperative period includes analyses of the vital signs, thorough physical examination with emphasis on the abdominal exam, laboratories including white blood cell count, radiologic studies such as upper GI study, and computed tomography (CT) scan of the abdomen and pelvis with PO and IV contrast. Upper endoscopy can play a role in defining the characteristics of a leak and may provide a therapeutic option in appropriate cir- a Fig. 17.1 (a) Upper GI contrast study demonstrating a leak from the proximal staple line following sleeve gastrectomy. (b) Computed tomography (CT) scan of the abdomen showing the staple line of the b sleeve gastrectomy with contrast extravasation proximally into an extraluminal collection (arrow) immediately adjacent to the gastric sleeve staple line 17 Management of Gastrointestinal Leaks and Fistula tinal leak is suspected, as reliance on false-negative imaging studies may delay operative intervention. Management Management of leaks after bariatric surgery varies and depends on the extent of the disruption, the abdominal contamination, the site of the leak, and timing of presentation. The initial treatments should include NPO (nothing per oral), early initiation of broad-spectrum antibiotics, and fluid resuscitation. Leaks can be classified as acute (within 7 days), early (1–6 weeks), late (6–12 weeks), and chronic (>12 weeks) [2]. The treatment options for postoperative leaks after bariatric surgery depend on the timing of presentation. Management of early and acute leaks can include nonoperative management, reoperation with abdominal washout with or without closure of the defect or placement of a T-tube in the defect, and wide peritoneal drainage. Alternatively, there have been few reports on the use of intraluminal stenting for gastric leaks after Roux-en-Y gastric bypass and sleeve gastrectomy [6–11]. For late and chronic leaks, management may require performing a proximal gastrectomy with esophagojejunostomy [12]. Nonoperative Treatment Nonoperative treatment can be considered for small, contained leaks, particularly in hemodynamically stable patients without peritoneal signs on abdominal exam. The patient should be kept NPO (nothing per oral) with total Fig. 17.2 Management strategies for staple line leak. Acute presentations can often be managed successfully with (a) insertion of intraluminal stent to cover the staple line defect or (b) drainage and T-tube insertion. (c) Chronic leaks from the staple line occasionally warrant resection of the involved area with Roux-en-Y reconstruction a 199 parenteral nutrition, and broad-spectrum intravenous antibiotic should be administered. The course for nonoperative treatment should be followed with measurement of serial white blood cell (WBC) count, vitals, and physical exam. Patients with increasing WBC, persistent tachycardia, or worsening of abdominal pain should proceed to endoscopic or operative management. In a relatively large study of leaks after gastric bypass, Gonzalez and colleagues reported successful nonoperative treatment in 23 of 26 patients, with an overall morbidity of 62% and no mortality [13]. Reoperation and Drainage The mainstay of surgical treatment includes drainage of all fluid collections and placement of abdominal drains with exclusion of the leak by endoluminal stent placement. Additionally, some surgeons make an attempt at closure of the defect; however, these closures tend to break down due to poor tissue integrity at the leak site. Leaks at the jejunojejunostomy or the gastric remnant may be more amenable for primary closure, and revision of the anastomosis is rarely needed [14]. An alternative approach to control the leak site is placement of a T-tube directly into the defect [10]. This technique consists of obtaining a conventional T-tube drain and placing directly into the defect (Fig. 17.2). The drain is then exteriorized and placed to bag drainage. The T-tube is left in place for 4–6 weeks and is slowly withdrawn over time (1–2 in. per week). The idea here is to create a track along the drain, thus creating a controlled fistula. Upon ­withdrawal of the tube, the well-formed fistulous track will collapse and eventually close. b 200 Fig. 17.2 (continued) N. T. Nguyen and S. C. Daly c Endoscopic Stent Endoscopic stenting for management of bariatric leaks was developed from the experience of using endoscopic stenting in management of esophageal anastomotic leaks after esophagectomy [15]. It is important to note that the use of endoscopic esophageal stent for management of leaks is an off-label use of a US Food and Drug Administration (FDA)approved device for malignant esophageal strictures or fistulas. Table 17.1 demonstrates selected series describing the use of endoscopic stent for treatment of gastric bypass or gastric sleeve leaks (Fig. 17.3) [6–11, 16]. To summarize the case studies, the use of endoluminal stents for the management of sleeve gastrectomy leaks has demonstrated a success rate of 55–100%. While stenting allows for early oral feeding during the healing process, stent migration, erosion, and intolerance remain risks of their use. Often, multiple interventions and stent exchanges are needed before complete healing is achieved [2]. Several principles should be followed when an esophageal stent is considered for management of a gastric leak after sleeve gastrectomy or gastric bypass. First, an endoscopy must be performed to evaluate the site of the leak, the size of the leak, and viability of the conduit. Second, appropriate drainage of abdominal collection is of utmost importance using either laparoscopic or percutaneous technique in combination with nothing per oral and nutritional support using either total parenteral nutrition or jejunostomy feeding. Third, the size of the endoscopic stent should be selected erring on the larger size in an effort to prevent migration. Endoscopic Stent Technique The procedure starts with an endoscopy to determine the site and extent of the leak. Upon confirmation of the site of leak, it is important to make sure that there is an adequate landing zone above the leak site. Additionally, it is important to estimate the luminal diameter of the esophagus in selection of the size and length of the stent. It is also important to use only fully covered stents rather than partially covered stents, which can lead to tissue ingrowth and possibly compromise its removal at a later date. There are two techniques for deployment of the esophageal stent, fluoroscopic guidance and endoscopic guidance. In the fluoroscopic guidance technique, the site of the leak is confirmed on fluoroscopy, and radiopaque markers (paper clips) are placed to outline the proximal and distal extent of the esophageal stent. An ultra-­ stiff guidewire is placed into the gastric antrum for sleeve gastrectomy patients or passed into the jejunal Roux limb for Roux-en-Y gastric bypass patients and confirmed under fluoroscopy. The endoscope is then removed, leaving the remaining guidewire in place. The selected esophageal stent is inserted transorally over the guidewire down the esophagus and positioned between the two radiopaque markers. The 17 Management of Gastrointestinal Leaks and Fistula 201 Table 17.1 Outcomes in management of leaks after selected series of gastric bypass and sleeve gastrectomy Authors/year/(reference) Sakran (2013) [12] Procedure (# patients Incidence of with leaks) leaks (%) Sleeve 44 De Aretxabala (2011) [18] Sleeve (n = 9) – Csendes (2010) [19] Sleeve (n = 16) 4.7% Tan (2010) [10] Sleeve (n = 14) – Casella (2009) [16] Sleeve (n = 6) 3.0% Freedman (2013) [20] Bypass (n = 69/2214) 3.1% Thodiyil (2008) [21] Bypass (n = 46) 1.7% Ballesta (2008) [22] Bypass (n = 59) 4.9% El Mourad (2013) [17] Sleeve (n = 26) Bypass (n = 18) – Spyropoulos (2012) [23] Sleeve (n = 12) Bypass (n = 5) BPD (n = 13) 2.0% Serra (2007) [7] Eisendrath (2007) [24] Sleeve or DS (n = 6) Bypass (n = 8) Sleeve gastrectomy (n = 4) DS (n = 8) BPD (n = 1) – – Eubanks (2008) [25] – Treatments Reop (n = 27) Perc drain (n = 28) Stent (n = 11) Lap reop (n = 5) Perc drain (n = 1) Open reop (n = 3) Stent (n = 4) Nonoperative (n = 8) Reop (n = 8) Nonoperative (n = 4) Reop (n = 7) Stent (n = 8) Perc drain (n = 5) Stent (n = 3) Nonoperative (n = 6) Stent (n = 35) Reoperation (n = 28) Reop (n = 27) Conservative (n = 33) Reop (n = 23) Conservative (n = 36) Stent (n = 41) Laparoscopy (n = 5) Endoscopic fibrin (n = 1) Stent (n = 9) Reop (n = 3) Nonoperative (n = 27) Stent (n = 6) Stent (n = 21) Reoperation (n = 3) Stent (n = 19) Resolution of leaks following stent insertion (%) Endoscopic stent with laparoscopic drainage Overall mortality 9.1% 75% 0% – 0% 50% 0% 100% 0% 100% 0% – 0% – 8.5% 96 0% 100% 3.3% 83% 81% 0% 19% 84% 0% Reop reoperation, Perc drain percutaneous drainage, BPD biliopancreatic diversion, DS duodenal switch stent is then deployed under fluoroscopic guidance. In the endoscopic guidance technique, the ultra-stiff guidewire is placed as above, and the endoscope is removed and then reinserted and positioned next to the guidewire at the level of the proximal aspect of stent deployment. The selected esophageal stent is inserted transorally over the guidewire under endoscopic guidance, and the stent is deployed. The endo- scope is used to ensure that the proximal aspect of the stent is positioned at the optimal position. Upon deployment of the stent, the guidewire is removed. A completion endoscopy is then performed with care to avoid dislodgement of the stent. In cases where migration is a high risk, the endoluminal stent may be either clipped or sutured into place at the proximal edge of the stent. N. T. Nguyen and S. C. Daly 202 present with a localized leak and without signs of systemic toxicity can include nonoperative management by rendering the patient NPO, providing parenteral nutrition and broad-­ spectrum antibiotic therapy, as well as percutaneously draining any intra-abdominal fluid collections. Endoscopic stenting has become an appropriate adjunct to this strategy and can obviate the need for reoperation for some patients. Surgical treatment including reoperation with drainage remains the mainstay of therapy for patients presenting with obvious signs of sepsis. In addition, chronic leaks and fistulae often require surgical reexploration with proximal gastrectomy and esophagojejunostomy. Question Section Fig. 17.3 Upper gastrointestinal contrast study showing a stent deployed for treatment of a proximal staple line leak after sleeve gastrectomy. Oral contrast passed entirely through the stent without evidence of contrast extravasation Chronic Fistula 1. Appropriate management for acute postoperative leaks includes all except: A. Endoluminal stent B. Laparoscopy with drainage C. Resection of the leak site and reconstruction D. IR drainage and IV antibiotic 2. In patients with suspected postoperative leaks, what is the first vital sign to be abnormal? A. Elevate systolic blood pressure B. Temperature greater than 101° C. Increase respiratory rate D. Tachycardia >100 Management of chronic fistula is often difficult due to the persistent leak into a well-contained cavity and often a well-­ defined epithelialized fistula tract. Initial management can References involve nonoperative strategies including endoscopic stent1. Khorgami Z, Shoar S, Andalib A, Aminian A, Brethauer SA, Schauer ing, endoscopic clipping, and/or injection of endoscopic PR. Trends in utilization of bariatric surgery, 2010–2014: sleeve fibrin glue. Failure of nonoperative management will require gastrectomy dominates. Surg Obes Relat Dis. 2017;13(5):774–8. surgical intervention such as surgical takedown of the fistula 2. Clinical Issues Committee of the American Society for Metabolic and Bariatric Surgery. Updated position statement on the prevention tract with proximal gastrectomy and esophagojejunostomy and detection of gastrointestinal leak. May 2015. [12]. Other authors have advocated using a Roux limb at the 3. Yehoshua RT, Eidelman LA, Stein M, Fichman S, Mazor A, Chen J, site of the chronic fistula [17]. Chronic leaks have also been et al. Laparoscopic sleeve gastrectomy–volume and pressure assessreported to result in a fistulous communication between the ment. Obes Surg. 2008;18(9):1083–8. 4. Hamilton EC, Sims TL, Hamilton TT, Mullican MA, Jones gastric lumen and pulmonary parenchyma or bronchus [12]. DB, Provost DA. Clinical predictors of leak after laparoscopic Management of leaks in this scenario may require lower lung Roux-en-Y gastric bypass for morbid obesity. Surg Endosc. lobectomy to ameliorate the fistula. 2003;17(5):679–84. Conclusion Gastrointestinal leak is a dreaded complication after bariatric surgery and is associated with significant morbidity and mortality. Early identification and management are very important in an effort to minimize the morbidity associated with the systemic inflammatory response and sepsis. A high index of suspicion is vital to facilitate expedient diagnosis and treatment of a staple line leak. Treatment of patients who 5. Warschkow R, Tarantino I, Folie P, Beutner U, Schmied BM, Bisang P, et al. C-reactive protein 2 days after laparoscopic gastric bypass surgery reliably indicates leaks and moderately predicts morbidity. J Gastrointest Surg. 2012;16(6):1128–35. 6. Nguyen NT, Nguyen X-MT, Dholakia C. The use of endoscopic stent in management of leaks after sleeve gastrectomy. Obes Surg. 2010;20(9):1289–92. 7. Serra C, Baltasar A, Andreo L, Pérez N, Bou R, Bengochea M, et al. Treatment of gastric leaks with coated self-expanding stents after sleeve gastrectomy. Obes Surg. 2007;17(7):866–72. 8. Simon F, Siciliano I, Gillet A, Castel B, Coffin B, Msika S. Gastric leak after laparoscopic sleeve gastrectomy: early covered self-­ expandable stent reduces healing time. Obes Surg. 2013;23:687–92. 17 Management of Gastrointestinal Leaks and Fistula 9. Oshiro T, Kasama K, Umezawa A, Kanehira E, Kurokawa Y. Successful management of refractory staple line leakage at the esophagogastric junction after a sleeve gastrectomy using the HANAROSTENT. Obes Surg. 2010;20(4):530–4. 10. Tan JT, Kariyawasam S, Wijeratne T, Chandraratna HS. Diagnosis and management of gastric leaks after laparoscopic sleeve gastrectomy for morbid obesity. Obes Surg. 2010;20(4):403–9. 11. Sakran N, Goitein D, Raziel A, Keidar A, Beglaibter N, Grinbaum R, et al. Gastric leaks after sleeve gastrectomy: a multicenter experience with 2,834 patients. Surg Endosc. 2013;27(1):240–5. 12. Sakran N, Assalia A, Keidar A, Goitein D. Gastrobronchial fistula as a complication of bariatric surgery: a series of 6 cases. Obes Facts. 2012;5(4):538–45. 13. Gonzalez R, Sarr MG, Smith CD, Baghai M, Kendrick M, Szomstein S, et al. Diagnosis and contemporary management of anastomotic leaks after gastric bypass for obesity. J Am Coll Surg. 2007;204(1):47–55. 14. Gonzalez R, Nelson LG, Gallagher SF, Murr MM. Anastomotic leaks after laparoscopic gastric bypass. Obes Surg. 2004;14(10):1299–307. 15. Nguyen NT, Rudersdorf PD, Smith BR, Reavis K, Nguyen X-MT, Stamos MJ. Management of gastrointestinal leaks after minimally invasive esophagectomy: conventional treatments vs. endoscopic stenting. J Gastrointest Surg. 2011;15(11):1952–60. 16. Casella G, Soricelli E, Rizzello M, Trentino P, Fiocca F, Fantini A, et al. Nonsurgical treatment of staple line leaks after laparoscopic sleeve gastrectomy. Obes Surg. 2009;19(7):821–6. 17. El Mourad H, Himpens J, Verhofstadt J. Stent treatment for fistula after obesity surgery: results in 47 consecutive patients. Surg Endosc. 2013;27:808–16. 203 18. de Aretxabala X, Leon J, Wiedmaier G, Turu I, Ovalle C, Maluenda F, et al. Gastric leak after sleeve gastrectomy: analysis of its management. Obes Surg. 2011;21(8):1232–7. 19. Csendes A, Braghetto I, León P, Burgos AM. Management of leaks after laparoscopic sleeve gastrectomy in patients with obesity. J Gastrointest Surg. 2010;14(9):1343–8. 20. Freedman J, Jonas E, Näslund E, Nilsson H, Marsk R, Stockeld D. Treatment of leaking gastrojejunostomy after gastric bypass surgery with special emphasis on stenting. Surg Obes Relat Dis. 2013;9(4):554–8. in press 21. Thodiyil PA, Yenumula P, Rogula T, Gorecki P, Fahoum B, Gourash W, et al. Selective nonoperative management of leaks after gastric bypass: lessons learned from 2675 consecutive patients. Ann Surg. 2008;248(5):782–92. 22. Ballesta C, Berindoague R, Cabrera M, Palau M, Gonzales M. Management of anastomotic leaks after laparoscopic Roux-­ en-­Y gastric bypass. Obes Surg. 2008;18(6):623–30. 23. Spyropoulos C, Argentou M-I, Petsas T, Thomopoulos K, Kehagias I, Kalfarentzos F. Management of gastrointestinal leaks after surgery for clinically severe obesity. Surg Obes Relat Dis. 2012;8(5):609–15. 24. Eisendrath P, Cremer M, Himpens J, Cadière G-B, Le Moine O, Devière J. Endotherapy including temporary stenting of fistulas of the upper gastrointestinal tract after laparoscopic bariatric surgery. Endoscopy. 2007;39(7):625–30. 25. Eubanks S, Edwards CA, Fearing NM, Ramaswamy A, la Torre de RA, Thaler KJ, et al. Use of endoscopic stents to treat anastomotic complications after bariatric surgery. J Am Coll Surg. 2008;206(5):935–8. Gastrointestinal Obstruction After Bariatric Surgery 18 Neil A. King and Daniel M. Herron Chapter Objectives 1. Address the evaluation of the most common presentations of partial and complete obstruction that occur after Roux-en-Y gastric bypass (RYGB), sleeve gastrectomy (SG), adjustable gastric band (AGB), and biliopancreatic diversion with duodenal switch (BPD-DS). 2. Explore management options for treating partial and complete obstructions following bariatric surgery. Introduction Obstruction After Roux-En-Y Gastric Bypass An understanding of the anatomy of RYGB is critical when diagnosing the bypass patient with obstruction, since the clinical presentation is determined by the location of the obstruction (Fig. 18.1). In RYGB, the gastric pouch is very small, 30 ml or less in volume and 5 cm or less in length. When the patient eats, food passes first through the pouch and then enters the Roux limb, a segment of jejunum typically 75–150 cm in length leading to the distal anastomosis. Food then passes through the “common channel”—the length of jejunum and ileum between the distal anastomosis and the ileocecal valve. The bypassed segment of the stomach, referred to as the “gastric remnant,” is no longer part of Obstruction of the gastrointestinal tract is one of the most common complications occurring after bariatric surgery. It may occur after any type of bariatric procedure and may range in severity from a minimal, spontaneously resolving problem to a life-threatening emergency. It is important to be familiar with the different types of strictures, partial obstructions, and complete obstructions that can occur postoperatively so that the patient can be expeditiously evaluated and appropriately treated. This chapter will address the evaluation and management of the most common presentations of partial and complete obstruction that occur after Roux-en-Y gastric bypass (RYGB), sleeve gastrectomy (SG), adjustable gastric band (AGB), and biliopancreatic diversion with duodenal switch (BPD-DS). N. A. King · D. M. Herron (*) Garlock Division of Surgery, Mount Sinai Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA e-mail: daniel.herron@mountsinai.org Fig. 18.1 Roux-en-Y gastric bypass © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_18 205 206 a N. A. King and D. M. Herron b Fig. 18.2 (a) This plain film of a gastric bypass patient with obstruction of the biliopancreatic limb is not particularly revealing. (b) CT imaging of the same patient reveals a tremendously enlarged gastric remnant filled with fluid. The antegastric Roux limb containing a small amount of contrast can be appreciated anteriorly the alimentary path but continues to secrete mucus and gastric acid. These gastric secretions join with bile and pancreatic fluid in the duodenum before passing though the ligament of Treitz into the biliopancreatic limb. The biliopancreatic limb is the bypassed segment of the intestine extending to the distal anastomosis. In general, obstruction of the Roux limb or common channel will result in obstructive symptoms familiar to the general surgeon, including nausea, vomiting, food intolerance, abdominal pain, and distention. Obstruction of the biliopancreatic limb is more challenging to diagnose, since the alimentary path may remain unblocked. In biliopancreatic limb obstruction, the gastric remnant may become severely distended with fluid yet remain invisible on plain film (Fig. 18.2a). This may result in abdominal fullness, bloating, hiccups, and pain but no nausea or vomiting. For this reason, computed tomography (CT) scan of the abdomen and pelvis is a critically important component of the evaluation of the patient with possible bowel obstruction, as it will reveal obstruction of both the bypassed and non-bypassed bowel (Fig. 18.2b). Obstruction of the biliopancreatic limb will result in severe distension of the gastric remnant and generally requires emergent decompression, either surgically or percutaneously. lowing gastric bypass varies from 0.9% to 34% [1–3]. A 2015 study including 1500 patients reported a gastrojejunal stricture rate requiring endoscopic intervention of 3.4% [4]. While a significant majority of gastrojejunal strictures present within the first 90 days after surgery, some patients may present much later, even a year or more postoperatively [5]. The typical stricture patient presents 4–6 weeks after surgery with solid food intolerance progressing to liquid intolerance as the stricture narrows. In severe cases, patients may be unable to swallow their own oral secretions. The diagnosis of stricture can usually be made based on history alone and confirmed with upper endoscopy. Radiographic contrast studies may or may not be helpful; they are not very sensitive in detecting strictures and pose the risk of contrast aspiration. Upper endoscopy is the primary diagnostic modality of choice, as it allows both rapid diagnosis and therapeutic intervention via balloon dilatation [6]. It is difficult to precisely measure a stricture, as no widely accepted definition of stricture exists, but most endoscopists consider an anastomosis that is too narrow to permit passage of a standard upper endoscope (approximately 9.5 mm in diameter) to represent a stricture. After being diagnosed endoscopically, the stricture can be immediately dilated using a through-the-scope dilating balloon up to a diameter of 12–15 mm. While most patients respond to a single dilatation, some will require a second or third; up to 13% may require four to five treatments [7]. Prior to the widespread use of dilating balloons, treatment of anastomotic stricture was achieved using bougie dilators like the Savary-Gilliard dilator (Wilson-Cook Medical Inc., Winston-Salem, NC). Some studies have demonstrated Gastrojejunal Stricture Stricture of the proximal anastomosis, or gastrojejunostomy, is one of the more common complications after gastric bypass. The reported incidence of gastrojejunal stricture fol- 18 Gastrointestinal Obstruction After Bariatric Surgery excellent results with this technique, with no serious complications and most patients requiring only one to two dilations [8, 9]. While balloon dilation is a safe and effective procedure, perforation may rarely occur. A 2012 review of 760 patients undergoing endoscopic dilation found perforation to be the most common complication occurring in 14 patients (1.8%). Of note, only two of these patients required operative intervention. The authors reported that only 15 patients (2%) required surgical revision for recurrent stenosis [10]. The etiology of gastrojejunal stricture formation is not well understood. Some believe it is related to ischemia, while others feel that suture material or staples may be causative. Stricture formation appears to be related to the initial size of the anastomosis; a 2012 study demonstrated a nearly fourfold incidence of intraluminal stenosis associated with the 21-mm circular stapler compared to the 25-mm circular stapler [11]. For stapled anastomoses, firm apposition or compression of the tissue edges may be helpful in reducing stricture rate. A recent study found that a circular stapler with a 3.5-mm height resulted in lower stricture rate than one with a 4.8-mm height (3.5 v 13.9%) [12]. The use of staple line reinforcement materials has also been shown to reduce stricture rate [13]. Some surgeons feel that handsewn anastomoses are less likely to stricture, while others prefer linear stapled or circular anastomosis. A meta-analysis of over 13,000 patients found no difference in stricture rates between handsewn and stapled anastomoses [14]. Although obstruction at the distal anastomosis is sometimes described as a “distal stricture,” these obstructions are most likely due to kinking of the anastomosis or excessive narrowing of the common enterotomy closure due to technical error. True stricture formation along a 60-mm stapled jejunojejunostomy anastomosis is exceedingly rare. Obstruction from Internal Hernia Internal hernias are relatively common after gastric bypass and may result in bowel obstruction, intestinal ischemia, or both. A recent study of 594 patients found internal hernia rates as high as 6.2% following Roux-en-Y gastric bypass [15]. A 2007 series from the Cleveland Clinic reported that internal hernia was the single most common cause of bowel obstruction in their gastric bypass patients, representing 41% of all obstructions [16]. In a gastric bypass patient with an antecolic Roux limb, two different types of internal hernias may potentially occur: • Distal anastomosis mesenteric hernia—this space is bordered by the divided mesentery of the biliopancreatic limb and the mesentery of the Roux limb at the distal anastomosis (Fig. 18.3). 207 Fig. 18.3 Internal hernia potential space at the mesenteric defect of the distal anastomosis • Petersen hernia—the space between the mesentery of the Roux limb and the mesocolon/colon (Fig. 18.4). Some bariatric surgeons perform a retrocolic gastric bypass in which the Roux limb is brought up behind the transverse colon through an opening in the mesocolon. These patients have the potential to suffer from a “mesocolic hernia” in which the Roux limb herniates up through the defect in the mesocolon through which the Roux limb passes. If the Roux limb is placed in an antecolic position, no such opening is created, and there is no potential for mesocolic herniation. Retrocolic gastric bypass is associated with a higher rate of internal hernias. A Mount Sinai study from 2003 found patients with retrocolic anastomoses to have nearly double the risk of developing an internal hernia (3.3% v 6.0%) [17]. While the majority of bariatric surgeons close these internal hernia spaces with permanent suture, some do not. A 2017 study found that closure of mesenteric defects resulted in a 2.5% internal hernia rate compared to nearly 12% in patients without mesenteric closure [18]. It is important to remember that even if hernia spaces have been sutured closed, hernia defects may still form at these sites. 208 N. A. King and D. M. Herron If the small bowel becomes entrapped within an internal hernia, its venous outflow may become partially or completely occluded, ultimately resulting in bowel ischemia and severe abdominal pain far out of proportion to the physical exam findings (Fig. 18.5). Typically, patients complain of intense pain in the midepigastrum, often radiating to the back. The pain may occasionally be relieved by leaning forward or getting “down on all fours,” maneuvers that serve to reduce the compression on the entrapped bowel. Entrapped bowel may reduce itself from the hernia spontaneously, bringing with it rapid resolution of symptoms, or it may persist until surgical intervention. Obstruction of the bowel lumen may or may not occur at the same time (Fig. 18.6). Physical exam is generally unrevealing in the patient with an internal hernia. Laboratory studies are similarly unhelpful. Computed tomography (CT) scan may demonstrate findings such as the “swirl sign,” a spiraling of the mesentery, mesenteric fat haziness, or intestinal distention in the upper abdomen [19]. However, these findings are neither sensitive nor specific, and it is essential to understand that a negative CT scan of the abdomen in no way rules out an internal hernia. Bowel obstruction may be absent even with a severe internal hernia causing intestinal ischemia. Thus, the gastric bypass patient with severe unrelenting abdominal pain but a normal CT scan still represents a surgical emergency. Fig. 18.4 Internal hernia potential space at between the Roux limb mesentery and the mesocolon, frequently referred to as a Petersen hernia Fig. 18.5 Petersen-type internal hernia after biliopancreatic diversion with duodenal switch, resulting in irreversible small bowel ischemia Fig. 18.6 Plain film of a patient with an incarcerated internal hernia, showing dilated small bowel loops. Bowel obstruction may or may not be present with internal hernia 18 Gastrointestinal Obstruction After Bariatric Surgery Definitive diagnosis of internal hernia can only be achieved through surgical exploration, either laparoscopic or open. If a significant length of bowel is entrapped within an internal hernia, intestinal anatomy may become so distorted that it becomes difficult to reduce the herniated bowel. In this situation, it is helpful to start by locating the ileocecal junction and then running the bowel in a retrograde fashion until the distal anastomosis is identified and the anatomy clarified. Once the herniated bowel is reduced, the hernia defect should be securely closed with a nonabsorbable running continuous suture. Some surgeons advocate the addition of a second layer of suture material or fibrin sealant to reinforce the hernia closure. If irreversibly ischemic bowel is identified, it will need to be resected; questionably viable bowel may warrant a “second-look” procedure within 24–48 h. mall Bowel Obstruction from Scars S and Adhesive Bands The surgeon should always remember that bariatric surgery patients are also general surgery patients and may suffer from intestinal obstruction due to intra-abdominal adhesions. In a review of bowel obstruction after gastric bypass, Elms et al. found that adhesions were the most common cause of bowel obstruction, accounting for 48% of the 93 patients that occurred in their series of 2395 patients [20]. Adhesions may form anywhere within the abdomen and can potentially obstruct the Roux limb, biliopancreatic limb, or common channel. Thus, plain films and even upper GI series may miss adhesive bowel obstructions that occur outside the alimentary path. CT scan with oral contrast is preferred since it will image the entire intra-abdominal gastrointestinal tract and may reveal the location of a transition zone where the obstruction is located. In the Cleveland Clinic series, plain films identified 35% of obstructions, while CT had a sensitivity of 51% [16]. If the Roux limb is placed in a retrocolic position at the time of surgery, it must necessarily pass through a surgically created defect in the mesocolon. This opening may potentially scar to the point of stricture formation. In the Cleveland Clinic series, this was the second most common cause of bowel obstruction in bypass patients, comprising 20% of all obstructions [16]. The risk of mesocolic stricture formation or mesocolic hernia can be completely eliminated at the time of initial surgery, if the surgeon places the Roux limb in an antecolic position. 209 hernia following laparoscopic Roux-en-Y gastric bypass [21]. The vast majority of these patients were asymptomatic, and only 2 of the 59 patients with ultrasound evidence of hernia required surgical repair during the study period. Elm’s review of 2395 laparoscopic bypass patients found just 1 case of incisional hernia requiring surgical repair [20]. To minimize the risk of incisional hernia, it is often recommended that surgeons close trocar sites 10 mm or larger. While this will not eliminate the risk of trocar site herniation entirely, it may significantly decrease the incidence [22]. The treatment of preexisting ventral hernias at the time of laparoscopic Roux-en-Y gastric bypass is more controversial. Most surgeons are reluctant to implant prosthetic mesh to repair large hernias during a clean-contaminated operation. Others feel that it is most prudent to wait until after the patient has achieved significant weight loss before performing the repair. Khorgami et al. reported a higher incidence of return to the operating room, readmission, discharge to rehabilitation facility, and a longer hospital length of stay among patients having concomitant ventral hernia repair and bariatric surgery when compared to patients having bariatric surgery alone [23]. Bariatric surgery with simultaneous mesh repair of ventral hernia does not yet constitute the standard of care. Intussusception Small bowel intussusception is a far less common cause of bowel obstruction after gastric bypass, with scattered case reports in the literature. The distal anastomosis can serve as a lead point for intussusception. If CT imaging can be obtained during the obstruction episode, the classic finding of the “target sign” will be pathognomonic for the condition (Fig. 18.7). Intussusception may or may not resolve spontaneously. If surgery is required, the intussusception should be reduced, with resection or revision of the presumed lead point if identifiable. Incisional Hernia Incisional hernias may form at open incisions or laparoscopic trocar sites or may exist as sequelae of earlier surgery. The true incidence of trocar site hernias may be underappreciated; a 2014 study using ultrasound to assess the abdominal wall found that 39% of patients developed a trocar site Fig. 18.7 CT imaging showing the classic finding of the “target sign” consistent with intussusception 210 bstruction from Intraluminal Blood Clot or O Bezoar Intraluminal causes of obstruction are exceedingly rare after gastric bypass. Koopman’s review in 2008 identified only ten reported cases [24]. Intraluminal blood clots, or hemobezoars, may form in the immediate postoperative period and can cause occlusion at the distal anastomosis. If the location of the bezoar is accessible endoscopically, the obstruction can be relieved nonsurgically. Otherwise, surgical exploration may be required with evacuation of the bezoar and confirmation of hemostasis. eneral Approach to the Bypass Patient G with Obstruction When approaching the gastric bypass patient with intestinal obstruction, it is important to remember that the clinical presentation is dependent upon the segment of bowel that is obstructed; an obstruction of the Roux limb will present far differently from an obstruction of the biliopancreatic limb. CT imaging with oral and IV contrast should be considered early in the diagnostic process as it will reveal obstruction in the Roux limb, biliopancreatic limb, and common channel. In contrast, plain films and upper gastrointestinal (UGI) series will generally be unhelpful in diagnosing obstruction of the bypassed bowel. Nasogastric tube decompression is frequently employed in management of the general surgical patient with bowel obstruction but is often unhelpful in the obstructed gastric bypass patient unless the blockage is extremely proximal and the patient is actively vomiting. Placement of a nasogastric tube may be harmful, as the tube can potentially disrupt a fresh gastrojejunal anastomosis or penetrate through the blind end of the Roux limb (“candy-cane end”) if forced. Finally, it should be repeated and emphasized that the patient with abdominal pain but no distended bowel on CT imaging may be suffering from an internal hernia. Internal hernias cause pain due to compression and ultimately obstruction of the intestinal vasculature and thus present with symptoms more suggestive of intermittent bowel ischemia than obstruction. N. A. King and D. M. Herron tive complications. Adjustable gastric bands are generally placed very high on the stomach, just beneath the gastroesophageal junction (Fig. 18.8). The band is designed to be appropriately sized for this location only and may cause excessive restriction or obstruction if it slips down to a wider area of the stomach or if the stomach prolapses up through the band. Bands are generally secured in place through a combination of natural anatomic factors and sutures placed specifically for this purpose. Most bands are placed using the “pars flaccida” technique, in which the posterior portion of the band is placed through the soft tissue of the retroperitoneum above the lesser sac. Because this portion of the band is not free in the intraperitoneal space, it is held in position by surrounding retroperitoneal soft tissue and surgical scar. In contradistinction, the anterior portion of the band is intraperitoneal and rests against the serosal surface of the anterior stomach. To secure this portion of the band, most surgeons imbricate the stomach over the band using simple interrupted or running sutures. Because gastric bands are located in the very proximal portion of the gastrointestinal tract, obstructions caused by them result in dramatic and easily recognizable symptoms. Patients present with nausea, vomiting, and the inability to tolerate solids or even liquids. In complete obstruction, the patient may even be unable to manage oral secretions. If the patient complains of epigastric pain, or is found to be tachycardic or febrile, the possibility of gastric ischemia should be considered and should prompt expeditious evaluation and bstruction in the Laparoscopic Adjustable O Gastric Band Patient The laparoscopic adjustable gastric band (LAGB) has significantly declined in popularity due to the emergence of data suggesting poor long-term outcomes and the availability of alternative operations such as sleeve gastrectomy [25, 26]. Nonetheless, hundreds of thousands of patients worldwide have bands in place, and it behooves both the general and bariatric surgeon to understand the band’s potential for obstruc- Fig. 18.8 Laparoscopic adjustable gastric band 18 Gastrointestinal Obstruction After Bariatric Surgery 211 management. Generally, the most appropriate first interven- anterior projection), a band will generally be viewed side-on. tion is to remove all fluid from the band. While this will not The left side of the band is normally somewhat higher than change the position of a slipped or prolapsed band, it may the right side; a line drawn through the body of the band is reduce the gastric obstruction enough to relieve symptoms, generally sloping upward to the left at 30–45° from the horiand its effect is immediate. zontal (Fig. 18.9). A plain film showing a completely flat If there is any question about the diagnosis, imaging of band suggests the possibility of anterior gastric slippage, the abdomen can be very useful to determine whether a band while a film showing a vertical band suggests posterior slipis in normal position. If a plain film is obtained (poster-­ page (Fig. 18.10). In the presence of gastric obstruction, an Fig. 18.9 (a) Plain film of the abdomen showing normal positioning of a gastric band. An upward angulation of the left side of the band of 30–45° is normal. (b) Esophagram showing normal positioning of a gastric band. This band is not yet filled and causes minimal holdup of contrast through the band. The band is angulated with the left side up about 30°, which is normal a b Fig. 18.10 Esophagram showing a slipped band with posterior gastric prolapse. Note the near-vertical orientation of the band and the excessively large stomach pouch 212 upper gastrointestinal series will quickly demonstrate lack of passage of contrast beyond the band. Care needs to be taken to avoid aspiration of contrast material in this setting, as this may result in severe aspiration pneumonitis. bstruction After Laparoscopic Adjustable O Gastric Band Placement The adjustable gastric band is designed to intentionally cause partial obstruction of the upper stomach. As a result, many gastric band patients report intermittent vomiting or regurgitation of food even when the band is appropriately positioned and adjusted. This “restrictive” effect can rapidly progress to complete obstruction if the band is aggressively overtightened or if it shifts out of position. Rapid recognition and intervention is essential to avoid gastric ischemia, necrosis, or perforation from an overly occlusive band. Early Postoperative Band Obstruction Gastric obstruction in the immediate postoperative period was not uncommon in the early days of the adjustable gastric band. A 2003 series of 56 patients described an 11% incidence of gastric obstruction within 24 h of band placement [27]. While three of the six obstructed patients in this series required immediate band removal, two others improved with conservative management, and one required surgery to reposition the band. The authors felt that the perigastric surgical technique they were using at the time resulted in a too low position on the stomach and reported a reduction in this complication after switching to the now-ubiquitous pars flaccida technique, which places the band just beneath the gastroesophageal junction. Newer bands are larger in diameter than earlier bands and allow a greater range of adjustment. The larger size, in conjunction with the use of the pars flaccida surgical technique for placement, has reduced the incidence of early obstruction [28]. Late Postoperative Band Obstruction Late obstruction may be caused by overly aggressive tightening of the band or by slippage of the band out of its original position. A review by Tice et al. found the incidence of band slippage to range from 1% to 20% [29]. The risk of band slippage persists during the patient’s lifetime, and the incidence will likely continue to increase as follow-up extends over longer periods. The band patient presenting with new obstructive symptoms should be queried regarding any recent adjustments. Regardless of the presumed cause of band obstruction, N. A. King and D. M. Herron removal of some or all of the fluid from the band is a prudent first step that may result in full resolution of symptoms. The band port should be accessed with a non-coring Huber-type needle if at all possible. However, if such a needle is not available, a narrow-gauge standard hypodermic needle can be used instead. If the access port cannot be localized by palpation alone, visualization of the port with fluoroscopy may be helpful. The patient should also be queried about the ingestion of food immediately prior to the obstructive symptoms; impacted food material may be easily removed with endoscopic intervention. The presence of epigastric or chest pain in addition to nausea or vomiting should raise the possibility of gastric ischemia; expedited workup and intervention is mandatory in this setting. If pain resolves immediately after deflation of the band, it may be safe to defer definitive therapy to a more elective setting. Persistent pain after band deflation warrants emergency reoperation. Similarly, the presence of fever or elevated white blood cell count suggests the possibility of gastric ischemia. In a 2006 report of 1275 banding patients, Ponce et al. described six different forms of band slippage [30]. Type I slippage involves prolapse of the posterior stomach through the band and is thought to be associated with the now-­ abandoned perigastric (intraperitoneal) band placement technique. Imaging will reveal a vertically positioned band (Fig. 18.10). Type II slippage describes anterior gastric prolapsed and may be associated with inadequate suture fixation of the band or excessive vomiting resulting in downward pressure on the device. Plain films may show a horizontally oriented band or one angulated downward to the left. Finally, type III slippage describes concentric pouch dilatation caused by excessive eating or an overly tight band and may be associated with a hiatal hernia or dilated esophagus [30]. Unusual Types of Band Obstruction Gastric bands may very occasionally cause obstruction by eroding into the lumen of the stomach or esophagus. Ruutiainen et al. described a patient whose band eroded completely into the stomach and migrated upward, causing obstruction at the gastroesophageal junction [31]. Alternatively, a fully eroded band may travel distally and occlude the small intestine. Operative intervention is required in these circumstances to remove the band and relieve the obstruction. It should be noted that erosion of the band through the gastric wall may occur without the emergence of sepsis or peritonitis. Additionally, it should be appreciated that this type of presentation is quite rare; erosion more ­commonly presents with sudden loss of restriction than with obstruction. Although uncommon, the tubing that connects the adjustable band to the subcutaneous port may cause bowel obstruc- 18 Gastrointestinal Obstruction After Bariatric Surgery tion. Several case reports have described this rare complication and its management [32, 33]. Surgical reduction of the bowel is required in this situation. Removal of the band, tubing, and port will eliminate the possibility of recurrence. Obstruction After Sleeve Gastrectomy Sleeve gastrectomy is now the most common bariatric operation in the United States. Generally, the operation involves resection of the greater curvature of the stomach with preservation of a banana-shaped stomach based on the lesser curvature (Fig. 18.11). Calibration of the stomach is accomplished by placing a bougie—typically 12–14 mm in diameter—along the lesser curvature during stapling. While the most feared and difficult-to-manage complication of the operation is gastric leakage, obstruction of the sleeve due to stenosis, twisting, or kinking can also present clinical challenges. It should be noted that many sleeve patients present with nausea and vomiting in the immediate postoperative period within 1–3 days after surgery. Upper GI series can be helpful to determine whether true mechanical obstruction is present. Patients with nausea and vomiting but without frank obstruction should be treated medically. Ondansetron and other antiemetic medications may be very helpful in relieving nausea, while some patients respond favorably to hyoscyamine, an anticholinergic. In our experience, some sleeve patients with Fig. 18.11 Laparoscopic sleeve gastrectomy 213 continued food intolerance without obstruction have responded favorably to the administration of mirtazapine, an antidepressant with beneficial gastrointestinal side effects. If upper GI series demonstrates true obstruction, endoscopic or surgical intervention is required. A 2013 review of 717 patients found symptomatic gastric stenosis in 5 patients (0.69%). Four of the five patients were successfully managed with endoscopic dilation, while one patient failed endoscopic treatment and underwent conversion to a Roux-en-Y gastric bypass [34]. bstruction After Biliopancreatic Diversion O with Duodenal Switch Biliopancreatic diversion with duodenal switch (BPD-DS) is a relatively uncommon bariatric/metabolic operation, less frequently performed than RYGB due to its greater technical complexity and short- and long-term morbidity. However, it has received some recent attention due to its extraordinary effectiveness with regard to both weight loss and metabolic improvement. A sleeve gastrectomy provides the restrictive component of BPD-DS. The duodenum is then divided just distal to the pylorus and anastomosed to the distal 250 cm of small intestine (Fig. 18.12). The remainder of the small bowel forms the biliopancreatic limb and is diverted from the alimentary tract. The biliopancreatic limb and the alimentary channel are anasto- Fig. 18.12 Biliopancreatic diversion with duodenal switch (BPD-DS) 214 mosed 50–100 cm from the ileocecal junction to form the common channel, where food and digestive juices intermix. Obstructive complication after BPD-DS are similar to those in gastric bypass, except that there is no gastric remnant present. Additionally, for reasons that are not clear, the risk of stenosis at the proximal anastomosis is much lower than that seen for gastric bypass. BPD-DS patients suffer from the same types of internal hernias as gastric bypass patients. The evaluation and management of a BPD-DS patient with abdominal pain or symptoms of obstruction are essentially identical to that of the bypass patient. N. A. King and D. M. Herron adhesions, incarcerated ventral hernias, inguinal hernias, etc. Thus, while we need to consider the many causes of bowel obstruction unique to bariatric operations, we should never forget that more conventional causes of obstruction may be present as well. As with general surgical patients, bariatric patients suffer significant morbidity and even mortality as a result of postoperative gastrointestinal obstruction. Hopefully, as knowledge regarding the evaluation and management of these obstructions is more widely disseminated, negative outcomes as a result of gastrointestinal obstruction will become increasingly rare. Obstruction from Implanted Medical Devices Many bariatric surgery patients suffer from gastroesophageal reflux disease (GERD) [35]. Magnetic sphincter augmentation was approved by the FDA in 2012 for use in treating GERD [36]. The LINX® Reflux Management System (Torax Medical, Shoreview, MN) is a magnetized beaded titanium band, which is placed laparoscopically around the ­gastroesophageal junction to control symptoms of reflux. Reports of treating GERD with the LINX® device in patients following bariatric surgery have been reported, with good outcomes [37, 38]. The LINX® device has been shown to have low complication or reoperation rates [39]. The most common reason for removal of these devices is severe dysphagia [39]; additionally isolated cases of the magnetic beads eroding into the esophagus have been reported in the literature [40, 41]. The LINX® device may play a larger role in treating bariatric patients with GERD in the future. The general surgeon should be aware of this novel technology and its potential risks and benefits. Intragastric balloons (IGB) are inflatable medical devices that are temporarily placed into the stomach to reduce weight. There are currently three devices that are approved by the FDA, the Orbera® (Apollo Endosurgery, Austin, TX), the ReShape Duo® (ReShape Medical, San Clemenete, CA), and the Obalon® (Obalon Therapeutics, Carlsbad, CA) [42]. Although uncommon, intragastric balloons can cause gastric outlet obstruction or can deflate and migrate causing distal small bowel obstruction. Gastric outlet obstructions can be treated with gastric decompression and endoscopic removal. Less than 1% of deflated balloons require surgical removal as most balloons will pass spontaneously from the rectum [43]. Conclusion Patients who have undergone bariatric surgery clearly present special concerns with regard to postoperative bowel obstruction. However, it is important to remember that bariatric patients are also general surgical patients and can suffer bowel obstruction from traditional sources such as Question Section 1. A patient presents to the emergency room 6 weeks after gastric bypass surgery. Although she was initially eating soft solid food without difficulty, she recently developed food intolerance that progressed to liquid intolerance as well. The most likely cause is: A. Obstruction from adhesions B. Internal hernia C. Intussusception D. Gastrojejunal stricture 2. A patient presents to the emergency room 2 years after Roux-en-Y gastric bypass complaining of severe unrelenting epigastric pain radiating to the back. The abdomen is only mildly tender on examination. Ultrasound and CT of the abdomen are unrevealing. The pain does not abate. The most appropriate next step is: E. Upper gastrointestinal series with small bowel follow-through F. HIDA scan G. Pain service consultation H. Surgical exploration 3. A patient with a history of adjustable gastric band placement 4 years ago presents with new-onset severe vomiting. A plain film shows the band to be vertically oriented. The best first step in managing this patient is: A. Withdraw all saline from the band B. Place a nasogastric tube C. Upper endoscopy D. Immediate surgical exploration References 1. Bell RL, Reinhardt KE, Flowers JL. Surgeon-performed endoscopic dilatation of symptomatic gastrojejunal anastomotic strictures following laparoscopic Roux-en-Y gastric bypass. Obes Surg Springer-Verlag. 2003;13(5):728–33. 2. Rossi TR, Dynda DI, Estes NC, Marshall JS. Stricture dilation after laparoscopic Roux-en-Y gastric bypass. Am J Surg Elsevier. 2005;189(3):357–60. 18 Gastrointestinal Obstruction After Bariatric Surgery 3. Dillemans B, Sakran N, Van Cauwenberge S, Sablon T, Defoort B, Van Dessel E, et al. Standardization of the fully stapled laparoscopic Roux-en-Y gastric bypass for obesity reduces early immediate postoperative morbidity and mortality: a single center study on 2606 patients. Obes Surg Springer-Verlag. 2009;19(10):1355–64. 4. Ribeiro-Parenti L, Arapis K, Chosidow D, Dumont JL, Demetriou M, Marmuse JP. Gastrojejunostomy stricture rate: comparison between antecolic and retrocolic laparoscopic Roux-en-Y gastric bypass. Surg Obes Relat Dis. 2015;11(5):1076–84. 5. Yimcharoen P, Heneghan H, Chand B, Talarico JA, Tariq N, Kroh M, et al. Successful management of gastrojejunal strictures after gastric bypass: is timing important? Surg Obes Relat Dis. 2012;8(2):151–7. 6. Boules M, Chang J, Haskins IN, Sharma G, Froylich D, El-Hayek K, et al. Endoscopic management of post-bariatric surgery complications. World J Gastrointest Endosc. 2016;8(17):591. 7. Ukleja A, Afonso BB, Pimentel R, Szomstein S, Rosenthal R. Outcome of endoscopic balloon dilation of strictures after laparoscopic gastric bypass. Surg Endosc Other Interv Tech Springer-­ Verlag. 2008;22(8):1746–50. 8. Fernández-Esparrach G, Bordas JM, Llach J, Lacy A, Delgado S, Vidal J, et al. Endoscopic dilation with Savary-Gilliard Bougies of stomal strictures after laparosocopic gastric bypass in morbidly obese patients. Obes Surg. 2008;18(2):155–61. 9. Escalona A, Devaud N, Boza C, Pérez G, Fernández J, Ibáñez L, et al. Gastrojejunal anastomotic stricture after Roux-en-Y gastric bypass: ambulatory management with the Savary–Gilliard dilator. Surg Endosc. 2007;21(5):765–8. 10. Campos JM, Salvo F, Mello T De, Antonio Á, Ferraz B, Nóbrega De Brito J, et al. Endoscopic dilation of gastrojejunal anastomosis after gastric bypass. ABCD Arq Bras Cir Dig. 2012;25:283–89. 11. Markar SR, Penna M, Venkat-Ramen V, Karthikesalingam A, Hashemi M. Influence of circular stapler diameter on postoperative stenosis after laparoscopic gastrojejunal anastomosis in morbid obesity. Surg Obes Relat Dis. 2012;8:230–5. 12. Horkoff M, Purich K, Switzer N, Prasad S, Church N, Shi X, et al. A shorter circular stapler height at the gastrojejunostomy during a Roux-En-Y gastric bypass results in less strictures and bleeding complications. J Obes. 2018;2018:1–4. 13. Scott JD, Cobb WS, Carbonell AM, Traxler B, Bour ES. Reduction in anastomotic strictures using bioabsorbable circular staple line reinforcement in laparoscopic gastric bypass. Surg Obes Relat Dis. 2011;7(5):637–42. 14. Jiang HP, Le LL, Jiang X, Qiao HQ. Meta-analysis of hand-sewn versus mechanical gastrojejunal anastomosis during laparoscopic Roux-en-Y gastric bypass for morbid obesity. Int J Surg Elsevier Ltd. 2016;32:150–7. 15. Al-Mansour MR, Mundy R, Canoy JM, Dulaimy K, Kuhn JN, Romanelli J. Internal hernia after laparoscopic antecolic Roux-­ en-­Y gastric bypass. Obes Surg. 2015;25(11):2106–11. 16. Rogula T, Yenumula PR, Schauer PR. A complication of Roux-­ en-­ Y gastric bypass: intestinal obstruction. Surg Endosc. 2007;21(11):1914–8. 17. Comeau E, Gagner M, Inabnet WB, Herron DM, Quinn TM, Pomp A. Symptomatic internal hernias after laparoscopic bariatric surgery. J Gastrointest Surg. 2012;16:641–5. 18. Aghajani E, Bent ·, Nergaard J, Leifson BG, Hedenbro J, Gislason Hjortur. The mesenteric defects in laparoscopic Roux-en-Y gastric bypass: 5 years follow-up of non-closure versus closure using the stapler technique. Surg Endosc 2017;31:3743–3748. 19. Ximenes MAS, Baroni RH, Trindade RMC, Racy MCJ, Tachibana A, Moron RA, et al. Petersen’s hernia as a complication of bariatric surgery: CT findings. Abdom Imaging. 2011;36(2):126–9. 20. Elms L, Moon RC, Varnadore S, Teixeira AF, Jawad MA. Causes of small bowel obstruction after Roux-en-Y gastric bypass: a review of 2,395 cases at a single institution. Surg Endosc. 2014;28:1624–8. 215 21. Scozzari G, Zanini M, Cravero F, Passera R, Rebecchi F, Morino M. High incidence of trocar site hernia after laparoscopic or robotic Roux-en-Y gastric bypass. Surg Endosc Other Interv Tech. 2014;28(10):2890–8. 22. Helgstrand F, Rosenberg J, Bisgaard T. Trocar site hernia after laparoscopic surgery: a qualitative systematic review. Hernia. 2011;15(2):113–21. 23. Khorgami Z, Haskins IN, Aminian A, Andalib A, Rosen MJ, Brethauer SA, et al. Concurrent ventral hernia repair in patients undergoing laparoscopic bariatric surgery: a case-matched study using the National Surgical Quality Improvement Program Database. Surg Obes Relat Dis. 2017;13(6):997–1002. 24. Koppman JS, Li C, Gandsas A. Small bowel obstruction after laparoscopic Roux-En-Y gastric bypass: a review of 9,527 patients. J Am Coll Surg. 2008;206(3):571–84. 25. Koh CY, Inaba CS, Sujatha-Bhaskar S, Hohmann S, Ponce J, Nguyen NT. Laparoscopic adjustable gastric band explantation and implantation at academic centers. J Am Coll Surg. 2017;225(4):532–7. 26. Chakravarty PD, McLaughlin E, Whittaker D, Byrne E, Cowan E, Xu K, et al. Comparison of laparoscopic adjustable gastric banding (LAGB) with other bariatric procedures; a systematic review of the randomised controlled trials. Surgeon. 2012;10:172–82. 27. Patel SM, Shapiro K, Abdo Z, Ferzli GS. Obstructive symptoms associated with the Lap-Band in the first 24 hours. Surg Endosc. 2004;18:51–5. 28. Nguyen NT, Hohmann S, Nguyen X-M, Elliott C, Masoomi H. Outcome of laparoscopic adjustable gastric banding and prevalence of band revision and explantation at academic centers: 2007– 2009. Surg Obes Relat Dis. 2012;8(6):724–7. 29. Tice JA, Karliner L, Walsh J, Petersen AJ, Feldman MD. Gastric banding or bypass? A systematic review comparing the two most popular bariatric procedures. Am J Med. 2008;121(10):885–93. 30. Ponce J, Fromm R, Paynter S. Outcomes after laparoscopic adjustable gastric band repositioning for slippage or pouch dilation. Surg Obes Relat Dis. 2006;2(6):627–31. 31. Ruutiainen AT, Levine MS, Dumon K. Intraluminal erosion and retrograde migration of laparoscopic gastric band with high-grade obstruction at gastroesophageal junction. Surg Obes Relat Dis. 2012;8(2):e14–6. 32. Hashemzadeh M, Karamirad M, Zahedi-Shoolami L. Laparoscopic adjustable gastric banding connecting tube causing small bowel obstruction and perforation. Case Rep Surg Hindawi Limited. 2013;2013:296037. 33. Strobos E, Antanavicius G, Josloff R. Unusual complication: small bowel obstruction caused by tubing of gastric band. Surg Obes Relat Dis. 2009;5(5):637–40. 34. Burgos AM, Csendes A, Braghetto I. Gastric stenosis after laparoscopic sleeve gastrectomy in morbidly obese patients. Obes Surg. 2013;23(9):1481–6. 35. Hajar N, Castell DO, Ghomrawi H, Rackett R, Hila A. Impedance pH confirms the relationship between GERD and BMI. Dig Dis Sci. 2012;57(7):1875–9. 36. Reynolds JL, Zehetner J, Bildzukewicz N, Katkhouda N, Dandekar G, Lipham JC. Magnetic sphincter augmentation with the LINX device for gastroesophageal reflux disease after U.S. Food and Drug Administration approval. Am Surg. 2014;80(10):1034–8. 37. Hawasli A, Tarakji M, Tarboush M. Laparoscopic management of severe reflux after sleeve gastrectomy using the LINX® system: technique and one year follow up case report. Int J Surg Case Rep Surg Assoc Ltd. 2017;30:148–51. 38. Desart K, Rossidis G, Michel M, Lux T, Ben-David K. Gastroesophageal reflux management with the LINX® system for gastroesophageal reflux disease following laparoscopic sleeve gastrectomy. J Gastrointest Surg. 2015;19(10):1782–6. 39. Lipham JC, Taiganides PA, Louie BE, Ganz RA, Demeester TR. Safety analysis of first 1000 patients treated with magnetic 216 sphincter augmentation for gastroesophageal reflux disease. Dis Esophagus. 2015;28(4):305–11. 40. Smith CD, Ganz RA, Lipham JC, Bell RC, Rattner DW. Lower esophageal sphincter augmentation for gastroesophageal reflux disease: the safety of a modern implant. J Laparoendosc Adv Surg Tech. 2017;27(6):586–91. 41. Bauer M, Meining A, Kranzfelder M, Jell A, Schirren R, Wilhelm D, et al. Endoluminal perforation of a magnetic antireflux device. Surg Endosc Other Interv Tech Springer US. 2015;29(12):3806–10. N. A. King and D. M. Herron 42. Laing P, Pham T, Taylor LJ, Fang J. Filling the void: a review of Intragastric balloons for obesity. Dig Dis Sci Springer US. 2017;62(6):1399–408. 43. Mathus-Vliegen EMH, Alders PRH, Chuttani R, Scherpenisse J. Outcomes of intragastric balloon placements in a private practice setting. Endoscopy. 2015;47(4):302–7. 19 Postoperative Bleeding in the Bariatric Surgery Patient Federico J. Serrot, Samuel Szomstein, and Raul J. Rosenthal Chapter Objectives 1. Classification of bleeding, etiologies, diagnosis, and management of bleeding in bariatric surgery patients. Introduction Bleeding is an uncommon complication in bariatric surgery. The reported incidence ranges between 0% and 4.4% and varies according to the different procedures performed [1–4] (gastric bypass (GBP), gastric banding, sleeve gastrectomy, and biliopancreatic diversion with duodenal switch). Bleeding after bariatric surgery can be classified based on time of onset (acute, early, late, and chronic) and based on bleeding site (intra-abdominal [IA] and intraluminal [IL]). The incidence of bleeding has decreased steadily due to the development of hemostatic agents (i.e., fibrin glue) and the use of staple-line reinforcement products, sophisticated stapling devices and energy sources, and the overall improvement of laparoscopic techniques. However, despite these advancements, bleeding remains a known complication that can be difficult to diagnose and manage in the bariatric surgery population. Failure to diagnose and manage bleeding in a timely manner can result in a significantly prolonged hospital stay and increased morbidity and mortality. F. J. Serrot Section of Minimally Invasive Surgery, Department of General Surgery, The Bariatric and Metabolic Institute Cleveland Clinic Florida, Weston, FL, USA S. Szomstein (*) · R. J. Rosenthal Department of General Surgery, The Bariatric and Metabolic Institute Cleveland Clinic Florida, Weston, FL, USA e-mail: szomsts@ccf.org Definition/Causes Bleeding, or hemorrhage, can be defined as the presence of hematemesis or melena and persistent large bloody output from a surgical drain, with or without the presence of tachycardia, hypotension, oliguria, and a decreasing hemoglobin and hematocrit [1]. Postoperative bleeding can be classified based on bleeding site into intraluminal bleeding (ILB, or into the gastrointestinal tract) or intra-abdominal bleeding (IAB, or into the abdominal cavity). ILB can occur anywhere in the gastrointestinal tract and can be classified based on time of onset into acute (1–7 days), early (1–6 weeks), late (6–12 weeks), and chronic (more than 12 weeks) (Table 19.1). Whereas IAB occurs mainly in the acute postoperative period and is near trocar sites, vascular pedicles, and/or staple lines, ILB can occur at any time and varies in etiologies from surgical reasons to medical reasons (peptic ulcer disease) or neoplastic (tumors) [5–7]. Acute or early postoperative bleeding usually occurs within the first 12–48 h after a surgical procedure but can still manifest as late as 42 days [5, 6]. In gastric sleeve patients, bleeding is mainly related to the long staple line, short gastric vessel pedicles, or trocar sites. In gastric bypass (GBP) patients, bleeding is usually related to an anastomotic or staple-line bleed as well as a stress ulcer or gastritis. The bleeding can be either IL or IA. Other potential causes of IAB in the acute phase can be related as mentioned to trocar sites, surgical dissection sites, or incomplete hemostasis of surgical sites prior to disinflation of the abdominal cavity. Late and chronic bleeding will usually present more than 42 days after the procedure, and it is mostly related to marginal ulceration, gastritis, or neoplasm. It can occur after any bariatric procedure but is most commonly seen with GBP Table 19.1 Postoperative bleeding classification based on time of onset Acute 1–7 days Early 1–6 weeks Late 6–12 weeks © Springer Nature Switzerland AG 2020 N. T. Nguyen et al. (eds.), The ASMBS Textbook of Bariatric Surgery, https://doi.org/10.1007/978-3-030-27021-6_19 Chronic >12 weeks 217 218 F. J. Serrot et al. patients due to the ulcerogenic nature of the gastrojejunal anastomosis. These ulcers can be seen anywhere at the gastrojejunostomy, in the excluded gastric remnant and duodenum. The etiologies of these ulcers are multifactorial, including local ischemia due to poor blood supply, anastomotic tension, the use of nonabsorbable sutures, increased gastric acidity, Helicobacter pylori infection, tobacco use, and nonsteroidal anti-inflammatory drug (NSAID) therapy [6, 7]. Chronic bleeding in the sleeve gastrectomy (SG) patient is uncommon but can be related to ulcers that can develop but are not as prevalent as in the GBP patient. Cases of chronic bleeding in gastric band (GB) patients have been reported in cases of band erosion, where patients can present with melena or hematemesis. Neoplasms that were not detected preoperatively or arise de novo after the bariatric procedure should be also kept in mind when a patient p­ resents with new onset of signs or symptoms of bleeding [8–11]. plications and tends to increase gradually and be cyclical in bariatric surgery patients with postoperative hemorrhage. This cyclical behavior of tachycardia in bleeding episodes differentiates from the septic pattern that ensues in patients with anastomotic leaks where tachycardia stays up and above 120 beats per minute without a cyclical pattern [7] (Fig. 19.1). In the acute setting, one of the biggest challenges is differentiating between IL and IA bleeding, as this will guide further decision-making. Clinical presentation can be helpful when symptoms are typical of IL bleeding like melena and hematemesis. When the symptoms are not present, the use of surgical drains in the immediate postoperative period can guide the surgeon to the appropriate diagnosis as mentioned by Chousleb et al. [2]. If the drains are filling up with bright red blood, it is safe to assume the bleeding is IA. IL bleeding is more commonly seen after laparoscopic Roux-en-Y gastric bypass (LRYGB) due to the many staple lines that are present (gastrojejunostomy, jejunojejunostomy, and gastric remnant) (Fig. 19.2). Heneghan et al. described that 40% of staple-line bleeds are from the gastric remnant, 30% from the gastrojejunostomy, and 30% from the jejunojejunostomy [12]. Nguyen et al. demonstrated that the most common site of bleeding is at the gastrojejunostomy [6]. Aside from clinical signs, important diagnostic modalities are upper endoscopy and computed tomography (CT). The latter is only an option when patients are clinically and hemodynamically stable. A CT scan can demonstrate collections/hematomas if bleeding is IA and sometimes show fluid in the gastric remnant, which is a sign of IL bleeding. CT scan with intravenous contrast can also potentially show, though rare, if there is active bleeding with the presence of a Diagnostic Algorithm Acute and Early Bleeding Diagnosis of hemorrhage in the acute postoperative period, the first 48 h, is a challenging task. The bariatric population’s body habitus and the new anatomy result in difficult clinical decision-making. Some of the common signs of acute bleeding are increased bloody output if drains are left behind at the time of the primary procedure, abdominal distension, and/or tachycardia, hypotension, and oliguria. Tachycardia has been shown to be a very important indicator of postoperative com- Heart Rate (bpm) a Tachycardia Pattern in Leak after LRYGB 140 120 100 80 60 40 20 0 0 20 40 60 80 100 120 140 160 140 160 Time elapsed after surgery (h) b Heart Rate (bpm) Fig. 19.1 Patterns of tachycardia after (a) leak is sustained >120 bpm, (b) bleeding is cyclical LRYGB laparoscopic Roux-en-Y gastric bypass Tachycardia Pattern in Bleeding after LRYGB 140 120 100 80 60 40 20 0 0 20 40 60 80 100 Time elapsed after surgery (h) 120 19 Postoperative Bleeding in the Bariatric Surgery Patient 219 biliopancreatic limb. A laparoscopic utility gastrotomy can also be performed to gain access to and evaluate the gastric remnant with an endoscope when a double-balloon endoscopy is not possible. If all of the above are negative, new modalities such as capsule endoscopy to identify unusual small bowel pathology can be helpful [20–22]. Treatment Strategies Acute and Early Fig. 19.2 Potential sites for bleeding after gastric bypass and sleeve gastrectomy blush/active extravasation. Upper endoscopy is a very useful diagnostic modality since an intervention can be done without delay at the bedside with the patient in the intensive care unit (ICU) to stop the bleeding. Endoscopy should be performed with caution in the immediate postoperative period to avoid disruption of the anastomosis [13–16]. A negative esophagogastroduodenoscopy (EGD) should prompt the surgeon about a possible bleeding site from the jejunojejunostomy or gastric remnant. Late and Chronic Bleeding Chronic postoperative bleeding (>30 days) is mostly IL and often presents with clinical signs of upper gastrointestinal bleeding. Upper endoscopy tends to be the diagnostic method of choice [17–19]. Double-balloon enteroscopy can be performed to assess the excluded stomach and duodenum that can be the site of an ulcer or neoplasm. This tends to be a tedious procedure for gastroenterologists because it is easy to get lost in the anatomy and difficult to access the excluded Treatment of acute postoperative hemorrhage begins with the clinical assessment. It is important to make sure that the patient is hemodynamically stable. When bleeding is suspected, the patient should be placed in a monitored unit. A Foley catheter should be in place to assure adequate fluid resuscitation, and type and screen and serial blood hemoglobin/hematocrit should be obtained to determine the trend of blood loss and the need for possible blood transfusion. If the patient is clinically stable, further tests such as EGD can be performed, but if there is hemodynamic instability that is not responding to resuscitation, immediate surgical intervention might be required. The majority (>80%) of acute postoperative bleeding will resolve with conservative management, without the need for another procedure or reoperation [6, 13, 14]. It is very important to differentiate IL bleeding from IA bleeding in the immediate postoperative period. IA bleeding rarely requires a reoperation; the incidence of bleeding has dramatically decreased recently with wide use of staple-line reinforcement during bariatric procedures and with the advancement of hemostatic dissection devices (ultrasonic scalpel/bipolar vessel sealant). In the event that the bleeding does not stop and reoperation is warranted, diagnostic laparoscopy is an excellent option. If during the exploration no obvious source of bleeding is identified, all staple lines should be carefully examined and oversewn if considered appropriate [1]. If IL bleeding is diagnosed, conservative management remains an acceptable option; however, if the patient fails conservative management or is unstable, an invasive approach is the next step. Upper endoscopy is not only an excellent diagnostic modality but also a good treatment option in patients with acute IL postoperative bleeding. An endoscope can be used to treat bleeding in the gastric pouch of GBP patients and, with the aid of a laparoscopic gastrotomy, has access to the excluded small bowel/stomach. Endoscopic treatment options range from heat to injections to hemostatic clips. In the acute setting, the use of a heat source to stop bleeding is not recommended due to potential disruption of a fresh anastomosis; if possible, clips should be used. Many experts recommend that all endoscopic interventions should be done under laparoscopic observation to mon- 220 F. J. Serrot et al. Fig. 19.3 Algorithm for diagnosis/treatment of acute/ early bleeding. EGD esophagogastroduodenoscopy, Dx and Tx diagnosis and treatment ACUTE/EARLY BLEEDING Intra-abdominal Intraluminal Stable EGD (Dx and Tx) Drain Output Clinical Judgement Unstable Laparoscopy/ Laparotomy w/wo Intraoperative EGD NEGATIVE Stable Observation Unstable Laparoscopy/ Laparotomy Gastrotomy and Endoscopy itor for perforation [15]. If bleeding persists despite endoscopic management, oversewing of the affected area will be the definitive option (Fig. 19.3). Late and Chronic Bleeding When bleeding occurs 42 days or more after the procedure, it is most likely IL and presents with signs and symptoms of an upper or lower gastrointestinal bleed. This bleeding is more likely seen with GBP than other bariatric procedures. Since the most likely cause is ulcer disease, medical treatment with proton-pump inhibitors is an acceptable initial treatment method. If bleeding persists despite medical treatment or if there is hemodynamic instability, endoscopy is the best treatment option [19]. Upper endoscopy is the treatment method of choice for bleeding ulcers in the gastric pouch. Rabl et al. reported an 80% success rate at controlling bleeding in the gastric pouch or the gastrojejunostomy [15]. However, other reports show a success rate of <70% [16, 17]. Gastric and duodenal ulceration from the bypassed segment is most commonly due to marginal ulcer and reported to have a 7% incidence rate, but other sources of bleed include bile gastritis, helical pylori infection, and gastric cancer [23]. When the ulcers are present in the gastric remnant or the excluded duodenum, more advanced endoscopic procedures are required. Double-­balloon enteroscopy is a good option; however, the ability to access the biliopancreatic limb and to treat the bleeding is very operator dependent and does not have a high success rate. A variability of limb length among surgeons and adhesions from prior surgeries are some of the potential reasons for an unsuccessful doubleballoon enteroscopy. Following this, tagged red blood cell scan should be considered if the diagnosis is still lacking [24, 25]. A technically easier but more invasive procedure is a laparoscopic-assisted transgastric endoscopy. Using this method, the endoscopist is directed directly into the gastric remnant and has full access to the rest of the excluded duodenum and small intestine [21, 22, 26]. In patients with uncontrolled bleeding or recurrent ulcer disease, a surgical intervention is required. The aim of surgery is to treat the problem and to prevent a recurrence. For this reason, the surgical option of choice for patients with GBP with recurrent ulcers, uncontrolled ulcer bleeding, or multiple ulcer locations is an excision of the gastrojejunal anastomosis with reconstruction of a new gastrojejunal anastomosis or remnant gastrectomy, depending on the site of bleeding. Angiography and embolization have been described for upper gastrointestinal bleeding; however, in GBP patients, if the left gastric artery is embolized, ischemia of the gastric pouch is likely. For this reason, angiography should be reserved for only select patients who are not good surgical candidates [19] (Fig. 19.4). 19 Postoperative Bleeding in the Bariatric Surgery Patient LATE/CHRONIC BLEEDING Intraluminal POSITIVE (Marginal ulcer) Endoscopic Tx PPIs EGD NEGATIVE (Gastric remanent/duodenal ulcer) Bleeding scan Angiography Gastrotomy/EGD Double balloon endoscopy (?Tx at same time) STABLE Medical Treatment IV PPIs UNSTABLE Laparoscopy/ Laparotomy Embolization Fig. 19.4 Algorithm for treatment/management of late/chronic bleeding. EGD Endoscopic esophagogastroduodenoscopy; Tx and PPIS treatment and proton-pump inhibitors Special Considerations Staple-Line Reinforcement 221 cantly longer [25]. Another randomized trial from Dapri et al. concluded that buttressing reduces hemorrhage; however, there was no difference in the incidence of a leak or other postoperative complications [3]. Albanopoulos and colleagues, however, did not observe a significant difference in their rate of postoperative bleeding between patients with staple-line suturing or reinforcing buttressing material after LSG. These investigators had a 6% and 0% complication rate (bleeding and leak) in their suturing and buttressing material arms, respectively, but this did not reach significance [29]. Recently, Mohamed et al. compared the use of unidirectional barbed suture material (group A) and no reinforcement (group B). As shown before operative time was significantly higher in the suture group. Leak occurred in eight cases (1.7%) in the no reinforcement group and none (0%) in the barbed suture; leak was significantly lower in group A (p = 0.008). Bleeding occurred in two patients (0.4%) in group A and in seven (1.5%) in group B (p = 0.178), with no statistically significant difference between both groups [30]. Thromboprophylaxis Obesity is an important risk factor for the development of venous thromboembolism (VTE), as well as being an indeStaple-line reinforcement (SLR) has been a popular topic of pendent predictor for recurrent thromboembolic disease. discussion among experts in bariatric surgery. SLR includes VTE remains a leading cause of mortality after bariatric surbuttressing with absorbable materials such as glycolide and gery. Laparoscopic surgery does induce a postoperative trimethylene carbonate copolymer, porcine small intestine hypercoagulable state, as does open GBP, but it does so to a submucosa, or bovine pericardium strips. Recently, reports lesser degree. Incidence of VTE ranges between 0.8% and have described the use of hemostatic agents like Floseal 3.4%, and super obesity (BMI >60) appears to have a close (bovine-derived gelatin matrix of human-derived thrombin relationship with incidence of VTE and pulmonary embomixed with other components) with excellent results in lism. Low molecular weight heparin (LMWH) use for decreasing postoperative bleeding and rates of leaks [25, 26]. thromboprophylaxis in bariatric surgery patients has been SLR in bariatric surgery was initially described by widely accepted in the USA and worldwide. Severe bleedShikora et al. in 2004, when they reviewed buttressing mate- ing complications are infrequent with LMWH prophylaxis rial to reduce hemorrhage and leakage following GBP. They and lower than using weight-adjusted heparin doses. concluded that SLR with bovine pericardial strips may Thromboprophylaxis is safe and advised in bariatric surgery decrease the risk of acute staple-line failures [27]. The use of patients, and LMWH is favored over the unfractionated hephemostatic agents in GBP patients has also been described arin [31, 32]. by Silecchia et al. to prevent leaks, bleeding, and internal Recent data compared the use of different VTE prophyherniation [26]. A meta-analysis by Sajid et al. showed that laxis protocols and the effects on bleeding and thrombotic SLR reduces operative time in GBP patients, anastomotic complications following bariatric surgery. As expected, those leak, and number of clips needed to control hemorrhage at patients not receiving any VTE prophylaxis had the highest the staple line. However, SLR did not show superiority in rate of thrombotic complications. Following multivariable terms of controlling staple-line bleeding [28]. logistic regression, after adjusting for other factors, the risk Buttressing has gained attention with the increased popu- difference in having transfusion among different regimen larity of LSG. Gantileschi et al. randomized patients to three groups was significant (p value = 0.02), with patients in the groups with different SLR: oversewing, buttressing with a post-op prophylaxis and pre-post groups being significantly polyglycolide acid and trimethylene carbonate, and staple-­ less likely to have transfusion than patients with only preop line roofing with a gelatin-fibrin matrix. They observed no prophylaxis. When comparing results among patients receivdifferences in complications, but oversewing took signifi- ing heparin vs enoxaparin vs mixed, they noticed that those 222 receiving mixed protocols were more likely to have transfusion than patients having only heparin or enoxaparin (4.07 vs 2.50 vs 0.94%, p value <0.0001) [33]. Patients on Long-Term Oral Anticoagulation There are more than one million patients in North America requiring long-term oral anticoagulation due to various medical conditions. Perioperative management of the chronically anticoagulated patient is challenging in the general ­population. Morbidly obese population poses even a greater challenge due to multiple associated comorbidities and an increased risk for a thromboembolic event. Special perioperative considerations for patients receiving chronic anticoagulation therapy should include the original indication for the treatment, the time period from the last thromboembolic event, urgency of the surgery, risk of thromboembolism and hemorrhage, consequences associated with bleeding, and the duration of postoperative bleeding. Mourelo et al. examined 1700 patients undergoing bariatric surgery with 25 of those on chronic anticoagulation. Twenty-one patients underwent LRYGB, with three of those having bleeding complications but only one requiring a reoperation. They concluded that bariatric surgery can be safely performed in patients on chronic anticoagulation therapy [34]. ariatric Surgery in Patients Who Refuse Blood B Transfusion Most bleeding after surgery occurs within the first 24 h after surgery, and more than half of patients with bleeding required blood transfusion. This poses a problem in patients who refuse blood transfusions, with the best-known group being Jehovah’s Witness patients. Refusal of blood transfusion by Jehovah’s Witnesses has been associated with increased perioperative mortality after cardiac surgery but has not been demonstrated after other elective surgeries. Kitahama et al. found a low-average estimated blood loss and acceptable perioperative morbidity rates in the bloodless surgery patients who underwent bariatric surgery within the setting of a multidisciplinary program. Some techniques to minimize blood loss are laparoscopic surgery, meticulous intraoperative hemostasis, SLR, topical hemostatic agents, hypotensive anesthesia, and additionally using pediatric blood collection tubes in the postoperative period. A multidisciplinary approach is very important with investigation and treatment of preoperative anemia and perioperative advice regarding blood conservation and the use of blood substitutes [35]. F. J. Serrot et al. Conclusion Postoperative bleeding in the bariatric population is becoming rare with advancements in surgical tools. If bleeding does occur, expedient diagnosis and management is vital. Bariatric patients are best treated by bariatric surgeons who are familiar with the operation and anatomy. Question Section 1. What is the initial diagnostic modality of choice in a patient 6 months status post-gastric bypass who presents with melena and weakness? A. CT scan B. MRI with IV contrast C. Diagnostic laparoscopy D. Upper endoscopy 2. What is/are the therapeutic options for patient who presents with a bleeding ulcer in the gastric remnant? A. Observation B. Double-balloon enteroscopy C. Laparoscopic-assisted transgastric endoscopy D. Exploratory laparotomy E. B and C References 1. Mehran A, Szomstein S, Rosenthal R. Management of acute bleeding after laparoscopic roux-en-Y gastric bypass. Obes Surg. 2003;13:842–7. 2. Chousleb E, Szomstein S, Podkmeni D, Soto F, et al. Routine abdominal drains after laparoscopic Roux-en-Y gastric bypass: a retrospective review of 593 patients. Obes Surg. 2004;14:1203–7. 3. Dapri G, Cadiere GB, Himpens J. Reinforcing the staple line during laparoscopic sleeve gastrectomy: prospective randomized clinical study comparing three different techniques. Obes Surg. 2010;20:462. 4. Dick A, Byrne TK, Baker M, Budak A, Morgan K. Gastrointestinal bleeding after gastric bypass surgery: nuisance or catastrophe? Surg Obes Relat Dis. 2010;6(6):643–7. 5. Gill RS, Whitlock KA, Mohamed R, Sarkhoush K, Birch DW, Karmali S. The role of upper endoscopy in treating postoperative complications in bariatric surgery. J Interv Gastroenterol. 2012;2(1):37–41. 6. Nguyen NT, Langoria M, Chalifoux S, Wilson SE. Gastrointestinal bleeding after laparoscopic gastric bypass. Obes Surg. 2004;14:1308–12. 7. Dillemans B, Sakran N, Van Cauwenberge S, Sablon T, et al. Standardization of the fully stapled laparoscopic Roux-en-Y gastric bypass for obesity reduces early immediate postoperative morbidity and mortality: a single center study on 2606 patients. Obes Surg. 2009;19:1355–64. 8. Bellorin O, Abdemur A, Sucandy I, Szomstein S, Rosenthal RJ. Understanding the significance, reasons, and patterns of abnor- 19 Postoperative Bleeding in the Bariatric Surgery Patient mal vital signs after gastric bypass for morbid obesity. Obes Surg. 2011;21:707–13. 9. Schauer PR, Ikramuddin S, Gourash W, Ramanathan R, Luketich J. Outcomes after laparoscopic roux-en-Y gastric bypass for morbid obesity. Ann Surg. 2000;232:515–29. 10. Abeles D, Kim JJ, Tarnoff ME, Shah S, Shikora SA. Primary laparoscopic gastric bypass can be performed safely in patient with BMI >or = 60. J Am Coll Surg. 2009;208:236–40. 11. Podnos YD, Jimenez JC, Wilson SE, Stevens CM, Nguyen NT. Complications after laparoscopic gastric bypass:a review of 3464 cases. Arch Surg. 2003;138:957–61. 12. Heneghan HM, Meron-Eldar S, Yenumula P, Rogula T, Brethauer SA, Schauer PR. Incidence and management of bleeding complications after gastric bypass surgery in the morbidly obese. Surg Obes Relat Dis. 2012;8(6):729–35. 13. Ferreira LE, Song LM, Baron TH. Management of acute postoperative hemorrhage in the bariatric patient. Gastrointest Endosc Clin N Am. 2011;21:287–94. 14. Bakhos C, Alkhoury F, Kyriakides T, Reinhold R, Nadzam G. Early postoperative hemorrhage after open and laparoscopic roux-en-y gastric bypass. Obes Surg. 2009;19:153–7. 15. Rabl C, Peeva S, Prado K, James AW, et al. Early and late abdominal bleeding after roux-en-y gastric bypass: sources and tailored therapeutic strategies. Obes Surg. 2011;21:413–20. 16. Jamil LH, Krause KR, Chengelis DL, Jury RP, et al. Endoscopic management of early upper gastrointestinal hemorrhage following laparoscopic roux-en-y gastric bypass. Am J Gastroenterol. 2008;103:86–91. 17. Fernandez-Esparach G, Bordas JM, Pellise M, Gimeno-Garcia AZ, et al. Endoscopic management of early GI hemorrhage after laparoscopic gastric bypass. Gastrointest Endosc. 2008;67:552–5. 18. Monkhouse SJW, Morgan JDT, Norton SA. Complications of bariatric surgery: presentation and emergency management-a review. Ann R Coll Surg Engl. 2009;91:280–6. 19. Braley SC, Nguyen NT, Wolfe BM. Late gastrointestinal hemorrhage after gastric bypass. Obes Surg. 2002;12:404–7. 20. Strodel WE, Knol JA, Eckhauser FE. Endoscopy of the partitioned stomach. Ann Surg. 1984;200:582–6. 21. Sinar DR, Flickinger EG, Park HK. Retrograde endoscopy of the bypassed stomach segment after gastric bypass surgery: unexpected lesions. South Med J. 1985;78:255–8. 22. Sakai R, Kuga R, Safatle-Ribeiro AV, et al. Is it feasible to reach the bypassed stomach after Roux-en-Y gastric bypass for morbid obesity? The use of double-balloon enteroscope. Endoscopy. 2005;37:566–9. 223 23. Rasmussen JJ, Fuller W, Ali MR. Marginal ulceration after laparoscopic gastric bypass: an analysis of predisposing factors in 260 patients. Surg Endosc. 2007;21:1090–4. 24. Kitamura R, Lee J, Katz LB. The management of GI bleeding after gastric bypass surgery. Int J Surg Res Pract. 2015;2:2. 25. Gentileschi P, Camperchioli I, D’ugo S, et al. Staple-line reinforcement during laparoscopic sleeve gastrectomy using three different techniques: a randomized trial. Surg Endosc. 2012;26(9):2623–9. 26. Silecchia G, Boru CE, Mouriel J, Rossi M, et al. The use of fibrin sealant to prevent major complications following laparoscopic gastric bypass: results of a multicenter, randomized trial. Surg Endosc. 2008;22(11):2492–7. 27. Shikora SA. The use of staple-line reinforcement during laparoscopic gastric bypass. Obes Surg. 2004;14:1313–20. 28. Sajid MS, Khatri K, Singh K, et al. Use of staple line reinforcement in laparoscopic gastric bypass surgery: a meta-analysis. Surg Endosc. 2011;25:2884–91. 29. Albanopoulos K, Alevizos L, Flessas J, et al. Reinforcing the staple line during laparoscopic sleeve gastrectomy: prospective randomized clinical study comparing two different techniques. Preliminary results. Obes Surg. 2012;22(1):42–6. 30. Hany M, Ibrahim M. Comparison between stable line reinforcement by barbed suture and non-reinforcement in sleeve gastrectomy: a randomized prospective controlled study. Obes Surg. 2018;28(8):2157–64. 31. Hamad GG, Choban PS. Enoxaparin for thromboprophylaxis in morbidly obese patients undergoing bariatric surgery: findings of the prophylaxis against VTE outcomes in bariatric surgery patients receiving enoxaparin (PROBE) study. Obes Surg. 2005;15:1368–74. 32. Becattini C, Agnelli G, Manina G, et al. Venous thromboembolism after laparoscopic bariatric surgery for morbid obesity: clinical burden and prevention. Surg Obes Relat Dis. 2012;8:108–15. 33. Altieri M, Yang J, Hajagos J, et al. Evaluation of VTE prophylaxis and the impact of alternate regimens on post-operative bleeding and thrombotic complications following bariatric procedures. Surg Endosc. 2018;32(12):4805–12. 34. Mourelo R, Kaidar-Person O, Fajnwaks P, Roa PE, et al. Hemorrhagic and thromboembolic complications after bariatric surgery in patients receiving chronic anticoagulation therapy. Obes Surg. 2008;18:167–70. 35. Kitahama S, Smith MD, Rosencrantz DR, Patterson EJ. Is bariatric surgery safe in patients who refuse blood transfusion? Surg Obes Relat Dis. 2013;9(3):390–4. Management of Marginal Ulcers 20 Richard M. Peterson and Jason W. Kempenich Chapter Objectives 1. To understand the pathophysiology and development of marginal ulcer formation in the setting of Roux-en-Y (RNY) gastric bypass 2. To understand the implications of marginal ulcer disease and the prevention and treatment strategies for this disease Introduction The increasing incidence of obesity across the globe has led to a significant rise in the number of bariatric procedures performed for weight loss [1]. Lapar