Amylin Peptide: An Association with Feed Intolerance in Preterm Neonates and Infants of Diabetic Mothers ---------------------------------------------------------------------------------------------------------- Dr Venkatesh Ramulu Kairamkonda ---------------------------------------------------------------------------------------------------------- A Dissertation Submitted to the Faculty of Medicine in Partial Fulfilment of the Requirements for the Degree of Doctor of Medicine at the University of Sheffield ---------------------------------------------------------------------------------------------------------- This Research is conducted at Departments of Neonatal Intensive Care Unit and Academic Unit of Reproductive and Developmental Medicine, Jessop Wing, Royal Hallamshire Hospital and Neonatal Intensive Care Unit, Doncaster General Hospital ---------------------------------------------------------------------------------------------------------- November 2012 ---------- Declaration I hereby declare that my dissertation entitled “Amylin peptide: An association with feed intolerance in preterm neonates and infants of diabetic mothers” is the result of my own work and includes nothing which is the outcome of work done in collaboration except as declared in the Preface and specified in the text. It is not substantially the same as any that I have submitted, or, is being concurrently submitted for a degree or diploma or other qualification at the University of Sheffield or any other University or similar institution except as declared in the Preface and specified in the text. I further state that no substantial part of my dissertation has already been submitted, or, is being concurrently submitted for any such degree, diploma or other qualification at the University of Sheffield or any other University of similar institution except as declared in the Preface and specified in the text. Where other sources of information have been used, they have been referenced and/or acknowledged. Dr Anton Mayer explored the role of amylin in critically ill paediatric patients with feed intolerance that provided the basis to investigate the role of amylin in neonatal population. He also contributed significantly towards this project as a study collaborator. Myself and Drs Robert Coombs and Robert Fraser helped formulate hypotheses and overall study methodology in the neonatal setting. As a principle investigator I was responsible for ethics application and coordination of the three studies at Jessop Wing, Royal Hallamshire Hospital. I was also responsible for arranging funding, consenting, blood collection, data collection and completion of case report forms, analyses, presentation and report writing. Dr Anjum Deorukhkar was the principle investigator responsible for ethics application and coordination of study C at the neonatal unit, Doncaster General Hospital. Neonatal nurses at Jessop Wing and Doncaster General Hospital routinely performed and documented prefeed gastric residual volumes. Blood samples were also collected by junior doctors and colleague registrars. Analytical expertise including sample size calculations for the individual study was provided by the statisticians at this University. All blood samples were 1 assayed by Mrs Christine Bruce, technician at the academic unit of reproductive and developmental medicine, Jessop Wing, Royal Hallamshire Hospital. Name : Dr Venkatesh Ramulu Kairamkonda Signature: 2 Date: November 2012 Acknowledgements I would like to thank my supervisor, Mr Robert Fraser, Reader at the University of Sheffield and Consultant Obstetrician, section of Reproductive & Developmental Medicine at Jessop Wing, Royal Hallamshire Hospital. His constant enthusiasm, assistance, guidance, patience and advice helped me to complete the project successfully. I am indebted to Dr Anton Mayer, Consultant Paediatric Intensivist, Royal Hallamshire Hospital, Sheffield and Dr Robert Coombs, Consultant Neonatologist, Jessop Baby Unit, Sheffield for their advice, encouragement, assistance and support towards the initiation and publication of this research. I thank Mrs Christine Bruce for her invaluable contribution in the analyses of the blood samples, but most of all for her continual encouragement and kindness. I would like to thank the infants and their parents whose trust and co-operation enabled this work to be completed. I would also like to thank the Clinical staff and Nursing staff of the Neonatal Intensive care unit at Jessop Baby Unit and Doncaster General Hospital for their patience and assistance. I would like to thank the Baby Fund at the Jessop Wing, Royal Hallamshire Hospital for providing the financial support for this project. Finally, I would like to thank my wife Manjula and daughter Coral for their encouragement, understanding and support. 3 Abstract Title: Amylin peptide: An association with feed intolerance in preterm neonates and infants of diabetic mothers Introduction: Delayed enteral nutrition due to feed intolerance is common in preterm infants and infants of mothers whose pregnancy was complicated by diabetes mellitus (IMDM). Amylin, a 37 amino-acid polypeptide hormone, is a potent inhibitor of gastric emptying that may play a role in the patho-physiology of feed intolerance in these infants. Aims and Objectives: To determine serum amylin levels (i) at birth (umbilical cord) and postnatal day 5 (Guthrie) in healthy preterm and term infants-Study A, (ii) at birth (umbilical cord) and postnatal day 5 (Guthrie) in preterm and term IMDM-Study B, and (iii) in preterm infants experiencing feed-intolerance (nTOL) and feed-tolerance (TOL)-Study C. Hypothesis: Serum amylin levels are raised in (i) IMDM and (ii) preterm infants with increased gastric residual volumes (GRV); which may explain their feed intolerance. Methods and Material: Blood samples were analysed for total amylin immunoreactivity using monoclonal antibody based sandwich immunoassay. Results: Serum amylin concentrations (median (interquartile range)) in healthy term infants at birth (n=138) and postnatal day 5 (n=14) were 6.10 (3.30-9.70) pmol/L and 5.65 (3.10-8.20) pmol/L respectively. Similarly, the amylin concentrations in healthy preterm infants at birth (n=43) and postnatal day 5 (n=25) were 4.60 (1.90–8.30) pmol/L and 6.9 (2.75–9.50) pmol/L respectively. The amylin concentrations were significantly raised in both term IMDM at birth [n=17, 34.30 (28.35-50.00) pmol/L, p<0.0001] and postnatal day 5 [n=4, 25.20 (22.20-48.75) pmol/L, p<0.0001] and preterm IMDM at birth [n=14, 32.0 (18.6544.27) pmol/L, p<0.0001] and postnatal day 5 [n=9, 23.4 (15.37-46.57) pmol/L, p<0.0001]. 4 The amylin concentration was significantly elevated in nTOL group [n=30, 47.9 (21.4-79.8) pmol/L, p<0.0001)] compared to TOL group [n=30, 8.7 (5.7-16) pmol/L]. Conclusions: Amylin by virtue of its inhibitory effect on gastric emptying may be responsible in delaying establishment of enteral nutrition in preterm infants and infants of mothers whose pregnancy was complicated by diabetes mellitus. 5 Outline of Dissertation The structure of this dissertation is based on the suggested MD dissertation by the University of Sheffield. Chapter 1 introduces to the research project, study rationale, hypotheses, aims and objectives. Chapters 2 to 6 present literature review relevant to the dissertation. Chapters 7 and 8 to 10 outline the project plan and describe in detail methodology, results, discussion and conclusions of individual study. Chapters 11 and 12 conclude the dissertation with emphasis on suggested future studies and work. Chapters 13 and 14 provide publications arising out of this project and appendices respectively. Finally, chapter 15 outlines bibliography relevant to the dissertation. 6 Table of Contents Chapter 1- Overview & Background............................................................. 22 1.1 Foreword.............................................................................................. 22 1.2 Study Rationale ................................................................................... 23 1.3 Study Hypotheses ................................................................................ 23 1.4 Study Aims........................................................................................... 24 1.5 Study Objectives .................................................................................. 24 1.5.1 Primary Objectives ........................................................................ 24 1.5.2 Secondary Objectives ................................................................... 24 Chapter 2- Literature Review: Amylin Peptide ............................................. 26 2.1 Introduction & Background................................................................... 26 2.2 Amylin Synthesis, Sources, Release & Metabolism ............................. 26 2.3 Amylin Structure, Gene & Relation to CGRP Superfamily ................... 30 2.4 Feto-Maternal Passage of Amylin ........................................................ 32 2.5 Synthetic Analogue ‘Pramlintide’ ......................................................... 32 2.6 Amylin Receptors & Signal Transduction Mechanisms ........................ 34 2.7 Amylin Receptor Antagonist................................................................. 35 2.8 Biological Functions of Amylin ............................................................. 35 2.9 Mechanisms of Amylin Action at Molecular level ................................. 39 2.10 History of Discovery of Amylin .......................................................... 42 2.11 Association of Amylin with Islet Amyloid Deposits in Diabetes ......... 43 2.12 Amyloidosis and Composition of Amyloid Deposits .......................... 43 2.13 Laboratory Assays ............................................................................ 48 Chapter 3- Literature Review: Gastric Emptying .......................................... 50 3.1 Anatomy and Physiology of Gastric Motor Function ............................ 50 3.2 Regulation of Gastric Emptying ........................................................... 51 3.3 Role of Other Gastrointestinal Peptides on Gastric Emptying .............. 52 3.4 Diabetes and its Effects on Gastric Emptying ...................................... 53 3.5 Methods of Measurement of Gastric Emptying in Humans .................. 55 3.6 Methods and Factors Influencing Gastric Emptying in Neonates ......... 58 7 Chapter 4- Literature Review: Feed Intolerance in Preterm Infants ............. 60 4.1 Scale of the Problem............................................................................ 60 4.2 Physiology of Gastric and Gastrointestinal Motility in Preterm Infants . 60 4.3 Definition of Feed Intolerance .............................................................. 61 4.4 Patho-Physiology of Feed Intolerance ................................................. 63 4.5 Management of Feed Intolerance ........................................................ 65 Chapter 5- Literature Review: Gestational Diabetes Mellitus ....................... 69 5.1 Definition of Gestational Diabetes Mellitus........................................... 69 5.2 Scale of the Problem............................................................................ 69 5.3 Physiology of Insulin Secretion ............................................................ 69 5.4 Physiology of Gestational Diabetes Mellitus ........................................ 70 5.5 Strategies to Prevent Fetal and Neonatal Hyperinsulinaemia .............. 70 Chapter 6- Literature Review: IMDM............................................................ 72 6.1 Scale of the Problem............................................................................ 72 6.2 Physiology of Fetal Hyperinsulaemia ................................................... 72 6.3 Feed Intolerance in IMDM.................................................................... 73 6.4 Proposed Patho-Physiology of Feed intolerance in IMDM ................... 74 Chapter 7- Overview of Project Plan ............................................................ 75 7.1 General Design .................................................................................... 75 7.2 General Study Configuration and Methodology ................................... 78 7.3 Ethics Consideration ............................................................................ 81 7.4 Data Handling and Record Keeping .................................................... 83 7.5 Benefits and Risks ............................................................................... 84 7.6 Funding ................................................................................................ 84 Chapter 8- Clinical Study A: Umbilical & Guthrie Amylin in Preterm and Term Infants 85 8.1 Introduction .......................................................................................... 85 8.2 Methodology ........................................................................................ 85 8.2.1 Study Design & Setting ................................................................. 85 8.2.2 Consent ......................................................................................... 85 8 8.2.3 Inclusion Criteria............................................................................ 85 8.2.4 Exclusion Criteria .......................................................................... 86 8.2.5 Study Procedure............................................................................ 86 8.2.6 Study End Point............................................................................. 87 8.2.7 Sample Collection & Storage ........................................................ 88 8.2.8 Sample Analysis ............................................................................ 88 8.2.9 Sample Size .................................................................................. 89 8.2.10 Statistical Analysis ..................................................................... 89 8.2.11 Data Collection & Case Report Form ......................................... 90 8.2.12 Study Summary ......................................................................... 90 8.3 Results ................................................................................................. 92 8.4 Discussion ......................................................................................... 116 8.5 Conclusions ....................................................................................... 119 Chapter 9- Clinical Study B: Umbilical and Guthrie Amylin in Preterm and Term IMDM 120 9.1 Introduction ........................................................................................ 120 9.2 Methodology ...................................................................................... 120 9.2.1 Study Design & Setting ............................................................... 120 9.2.2 Consent ....................................................................................... 120 9.2.3 Inclusion Criteria.......................................................................... 120 9.2.4 Exclusion Criteria ........................................................................ 120 9.2.5 Study Procedure.......................................................................... 121 9.2.6 Study End Point........................................................................... 121 9.2.7 Sample Collection & Storage ...................................................... 121 9.2.8 Sample Analysis .......................................................................... 121 9.2.9 Sample Size ................................................................................ 121 9.2.10 Statistical Analysis ................................................................... 122 9.2.11 Data Collection & Case Report Form ....................................... 122 9.2.12 Study Summary ....................................................................... 122 9.3 Result................................................................................................. 125 9.4 Discussion ......................................................................................... 144 9.5 Conclusions ....................................................................................... 147 9 Chapter 10- Clinical Study C: Amylin Levels in Preterm infants with Feed Intolerance 148 10.1 Introduction ..................................................................................... 148 10.2 Methodology ................................................................................... 148 10.2.1 Study Design & Setting ............................................................ 148 10.2.2 Consent.................................................................................... 148 10.2.3 Inclusion Criteria ...................................................................... 148 10.2.4 Exclusion Criteria ..................................................................... 149 10.2.5 Study Procedure ...................................................................... 149 10.2.6 Definition of feed intolerance.................................................... 150 10.2.7 Definition of feed tolerance ...................................................... 151 10.2.8 Study End Point ....................................................................... 151 10.2.9 Sample Collection & Storage ................................................... 151 10.2.10 Sample Analysis ...................................................................... 151 10.2.11 Sample Size ............................................................................. 151 10.2.12 Statistical Analysis ................................................................... 151 10.2.13 Data Collection & Case Report Form ....................................... 152 10.2.14 Study Summary ....................................................................... 152 10.3 Results............................................................................................ 155 10.4 Discussion ...................................................................................... 166 10.5 Conclusions .................................................................................... 173 Chapter 11- Conclusions of the Dissertation................................................ 174 Chapter 12- Implications for Future Research ............................................. 176 Chapter 13- Publications arising from this Project ....................................... 179 13.1 Amylin peptide levels are raised in infants of diabetic mothers. Arch Dis Child. 2005 Dec; 90(12):1279-82........................................................... 179 13.2 Amylin peptide is increased in preterm neonates with feed intolerance. Arch Dis Child Fetal Neonatal Ed. 2008 Jul; 93(4):F265-70 ..... 184 Chapter 14- Appendices .............................................................................. 190 Chapter 15- Bibliography ............................................................................. 228 10 Key to Figures Figure 1. Amylin is the second beta cell hormone............................................. 26 Figure 2. Twenty four hour plasma profile of amylin and insulin in healthy adults .......................................................................................................................... 28 Figure 3. Amylin response after a liquid sustacal meal in patients with type 1 & type 2 diabetes mellitus .................................................................................... 29 Figure 4. Representative concentration profiles of amylin (A), total amylin immunoreactivity (TAI) (B) and insulin (C) ........................................................ 30 Figure 5. Alpha helix structure of human amylin ............................................... 31 Figure 6. Comparison of aminoacid sequence of amylin and pramlintide ......... 33 Figure 7. Role of amylin in regulation of postprandial glycemia ........................ 36 Figure 8. Amylin binding sites in rat brain ......................................................... 39 Figure 9. Location of amylin action within circumventricular organs in rat brain 41 Figure 10. Amyloid deposit in pancreatic islet ................................................... 44 Figure 11. Amyloid deposit in lymph node showing birefringence..................... 45 Figure 12. Extended cross-beta structure of amyloid ........................................ 46 Figure 13. Superpleated beta structure of amyloid fibrils .................................. 47 Figure 14. Region responsible for amyloid formation ........................................ 48 Figure 15. Site of synthesis of main gut hormones influencing appetite and gastric emptying ................................................................................................ 53 Figure 16. Project plan Gantt chart ................................................................... 75 Figure 17. Study plan Gantt chart ..................................................................... 79 Figure 18. General study methodology ............................................................. 80 11 Figure 19. Study A Gantt chart.......................................................................... 92 Figure 20. Study A flow diagram of participants and blood samples ................. 93 Figure 21. Box-and-whisker plot of umbilical cord amylin levels in healthy term infants ............................................................................................................... 96 Figure 22. Box-and-whisker plot of Guthrie amylin levels in healthy term infants .......................................................................................................................... 98 Figure 23. Notched box-and-whisker plot of amylin levels at birth and postnatal day 5 in healthy term infants ........................................................................... 100 Figure 24. Box-and-whisker plot of umbilical cord amylin levels at birth in healthy preterm infants ................................................................................... 102 Figure 25. Box-and-whisker plot of amylin levels at postnatal day 5 in healthy preterm infants ................................................................................................ 104 Figure 26. Notched box-and-Whisker plot of amylin levels at birth and postnatal day 5 in healthy preterm infants ...................................................................... 106 Figure 27. Comparison Box-and-Whisker plot of amylin level in term and preterm infants at birth .................................................................................... 108 Figure 28. Comparison Box-and-Whisker plot of serum amylin level in term and preterm infants on postnatal day 5 .................................................................. 109 Figure 29. The correlation graph showing line of best fit to assess the degree of association between paired umbilical cord venous amylin (x axis) and umbilical cord arterial amylin (Y axis)............................................................................. 110 Figure 30. Bland Altman method comparison plot of paired venous and arterial levels ............................................................................................................... 111 Figure 31. Bland Altman method comparison, percentage difference plot, of paired venous and arterial levels .................................................................... 112 12 Figure 32. The correlation graph showing line of best fit to assess the degree of association between paired umbilical cord insulin (x axis) and umbilical cord amylin (Y axis) in healthy preterm and term infants ........................................ 114 Figure 33. The correlation graph showing line of best fit to assess the degree of association between gestational age at birth (x axis) and umbilical cord amylin (Y axis) ............................................................................................................ 115 Figure 34. The correlation graph showing line of best fit to assess the degree of association between birth weight (x axis) and umbilical cord amylin (Y axis) . 116 Figure 35. Study B Gantt chart........................................................................ 124 Figure 36. Study B-flow diagram of participants and blood samples............... 125 Figure 37. Box-and-whisker plot of umbilical cord amylin levels in term IMDM infants ............................................................................................................. 127 Figure 38. Box-and-whisker plot of Guthrie amylin levels in term IMDM infants ........................................................................................................................ 129 Figure 39. Comparison Box-and-Whisker plot of amylin levels at birth and postnatal day 5 in term IMDMs ....................................................................... 131 Figure 40. Box-and-whisker plot of umbilical cord amylin levels in preterm IMDM infants ............................................................................................................. 133 Figure 41. Box-and-whisker plot of Guthrie amylin levels in preterm IMDM infants ............................................................................................................. 135 Figure 42. Comparison box-and-whisker plot of amylin concentration at birth and postnatal day 5 in preterm IMDM ............................................................. 137 Figure 43. Comparison box-and-whisker plot of amylin concentration in umbilical cord and Guthrie blood in term and preterm IMDM .......................... 138 Figure 44. Comparison box-and-whisker plot of amylin levels at birth and postnatal day 5 between term IMDM and healthy term infants (controls) ....... 140 13 Figure 45. Comparison box-and-whisker plot of amylin levels at birth and postnatal day 5 between preterm IMDM and healthy preterm infants (controls) ........................................................................................................................ 142 Figure 46. The correlation graph showing line of best fit to assess the degree of association between umbilical cord insulin (x axis) and umbilical cord amylin (Y axis) from paired umbilical cord blood samples of healthy and IMDM preterm and term infants .............................................................................................. 143 Figure 47. Study C Gantt chart ....................................................................... 154 Figure 48. Study C-Flow diagram of participants and blood samples ............. 155 Figure 49. Box and whisker plot of amylin levels (y-axis) in feed-intolerant (nTOL) and feed-tolerant (TOL) neonates (x-axis) .......................................... 158 Figure 50. Box and whisker plot of percentage GRV (y axis) in feed-intolerant (nTOL) and feed-tolerant (TOL) neonates (x axis) .......................................... 159 Figure 51. The box and whisker plot of days to reach full feeds (Y axis) in feedintolerant (nTOL) and feed-tolerant (TOL) neonates (X axis) .......................... 160 Figure 52. Correlation graph showing line of best fit to assess the degree of association between amylin concentration (y axis) and % GRV (x axis) in feedintolerant (nTOL) group ................................................................................... 161 Figure 53. The correlation graph showing line of best fit to assess the degree of association between amylin concentration (y axis) and days to reach full enteral feeds (x axis) in nTOL group ........................................................................... 162 Figure 54. The correlation graph showing line of best fit to assess the degree of association between days to discharge (x axis) and amylin concentration (Y axis) in the feed-intolerant (nTOL) group ........................................................ 163 Figure 55. The correlation graph showing line of best fit to assess the degree of association between % GRV (x axis) and amylin levels (y axis) in feed-tolerant (TOL) group .................................................................................................... 164 14 Figure 56. The correlation graph showing line of best fit to assess the degree of association between days to full enteral feeds (x axis) and amylin levels (Y axis) in feed-tolerant (TOL) group ........................................................................... 165 Figure 57. The correlation graph showing line of best fit to assess the degree of association between days to discharge (x axis) and amylin levels (Y axis) in the feed-tolerant (TOL) group ............................................................................... 166 Figure 58. Proposed pathway of amylin action ............................................... 168 15 Key to Tables Table 1. Study Objectives & Outcome Variables .............................................. 25 Table 2. Study A Summary ............................................................................... 91 Table 3. Summary statistic of amylin concentration and demographic characteristics at birth in healthy term infants ................................................... 95 Table 4. Summary statistic of amylin concentration and demographic characteristics at postnatal day 5 in healthy term infants .................................. 97 Table 5. Demographic characteristics of healthy term infants at birth and postnatal day 5.................................................................................................. 99 Table 6. Summary statistic of amylin concentration and demographic characteristics at birth in healthy preterm infants ............................................ 101 Table 7. Summary statistic of amylin concentration and demographic characteristics at postnatal day 5 in healthy preterm infants ........................... 103 Table 8. Demographic charateristics of healthy preterm infants at birth and postnatal day 5................................................................................................ 105 Table 9. Comparison of demography and amylin levels in healthy term and preterm infants at birth and postnatal day 5 .................................................... 107 Table 10. Study B summary ............................................................................ 123 Table 11. Summary statistic of amylin concentration and demographic characteristics at birth in term IMDM infants ................................................... 126 Table 12. Summary statistic of amylin concentration and demographic characteristics on postnatal day 5 in term IMDM infants ................................. 128 Table 13. Demographic characteristic of term IMDM at birth and postnatal day 5 ........................................................................................................................ 130 16 Table 14. Summary statistic of amylin concentration and demographic characteristics at birth in preterm IMDM infants .............................................. 132 Table 15. Summary statistic of amylin concentration and demographic characteristics at postnatal day 5 in preterm IMDM infants ............................. 134 Table 16. Demographic characteristics of preterm IMDM at birth and postnatal day 5 ............................................................................................................... 136 Table 17. Demographic characteristics and serum amylin levels of term healthy controls and term IMDM at birth and postnatal day 5 ...................................... 139 Table 18. Demographic characteristics and serum amylin levels of preterm healthy controls and preterm IMDM at birth and postnatal day 5 .................... 141 Table 19. Study C summary............................................................................ 153 Table 20. Baseline characteristics at the time of blood sampling of preterm neonates defined as feed-intolerant (nTOL) and feed-tolerant (TOL) ............. 156 Table 21. Serum amylin levels, % GRV, days to reach full feeds and length of stay in feed-intolerant (nTOL) and feed-tolerant (TOL) neonates ................... 157 17 Key to Appendices Appendix 1.Basic principle of luminescent ELISA ........................................... 190 Appendix 2. Human Amylin (Total) ELISA KIT and Assay Procedure ............. 193 Appendix 3. Characteristics of Human Amylin (Total) ELISA Kit..................... 196 Appendix 4. Research protocol for ethics approval, South Sheffield Research Ethics Committee ............................................................................................ 199 Appendix 5. Research protocol for ethics approval, Doncaster Research Ethics Committee....................................................................................................... 205 Appendix 6. Child participation information leaflet (Study A & B) .................... 210 Appendix 7. Child participation information leaflet (Study C) Royal Hallamshire Hospital ........................................................................................................... 213 Appendix 8. Child participation information leaflet (Study C) Doncaster Royal Infirmary .......................................................................................................... 216 Appendix 9. Study A & B Consent form .......................................................... 219 Appendix 10. Study C Consent form ............................................................... 220 Appendix 11. Advert for Study A ..................................................................... 221 Appendix 12. Advert for Study B ..................................................................... 222 Appendix 13. Advert for Study C ..................................................................... 223 Appendix 14. Case report form Study A .......................................................... 224 Appendix 15. Case report form Study B .......................................................... 225 Appendix 16. Case report form Study C ......................................................... 226 18 Key to Abbreviations AC 66 Amylin antagonist, also known as salmon calcitonin AC187 Amylin antagonist ACSA Antral cross sectional area ADP Adenosine diphosphate AMY 1, AMY 2 and AMY 3 Amylin receptors ATP Adenosine triphosphate BMI Body Mass Index CPAP Continuous positive airway pressure CT Calcitonin CTR Calcitonin receptor CGRP Calcitonin gene related peptide CV Coefficient of variation C.F.U Colony forming unit CCK Cholecystokinin CRF Case Report Form DBS Dried blood spot 19 EGG Electrogastrography ELISA Enzyme linked Immunosorbent Assay ELBW Extremely low birth weight infants FDA Food and drug administration, USA 4-MUB 4-Methylumbelliferyl Phosphate GDM Gestational diabetes mellitus GIP Glucagon inhibitory peptide GLP-1 Glucagon like peptide 1 GLP-2 Glucagon like peptide 2 GRV Gastric residual volumes GLUT 1 and GLUT 2 Glucose transporters GSK 3 Glycogen synthase kinase 3 IGF Insulin like growth factor IGFBP Insulin like growth factor binding protein IMDM Infant of mothers whose pregnancy was complicated by diabetes mellitus IAPP Islet amyloid polypeptide KATP ATP sensitive potassium channel 20 MMC Migrating motor complex NEC Necrotising enterocolitis nTOL Preterm infants with feed intolerance PC2 Protein convertase 2 PK-PD Pharmacokinetic-Pharmacodynamic PYY Peptide YY RAMP Receptor activity modifying proteins RFU Relative Fluorescent Unit RIA Radioimmunoassay sCT Salmon calcitonin, amylin antagonist TBS Tris Buffered Saline TOL Preterm infants without intolerance TPN Total parenteral nutrition VLBW Very low birth weight infants 21 feed Chapter 1 - Overview & Background Chapter 1- Overview & Background 1.1 Foreword Neonatologists often struggle to attain full enteral nutrition in majority of preterm infants with very low birth weight due to common occurrence of feed intolerance (Reddy, Deorari et al. 2000; ElHennawy, Sparks et al. 2003; Patole, Rao et al. 2005) characterised by large gastric residual volumes (GRV) with or without associated clinical manifestations (Jadcherla and Kliegman 2002; Mihatsch, von Schoenaich et al. 2002; Patole 2005). Consequently feeds are either reduced or withheld and frequently these neonates are subjected to investigations and treatments for presumed necrotising enterocolitis (NEC). Reliance on total parenteral nutrition (TPN) during this period is usually not enough to maintain protein and lipid intake (Grover, Khashu et al. 2008; Hay 2008) and this often leads to problems with infection and hepatic damage (Donnell, Taylor et al. 2002; Kaufman, Gondolesi et al. 2003). These infants may lag behind in protein and calorie intake by up to 6-8 weeks by the time they are ready to go home (Grover, Khashu et al. 2008) which may have adverse effects on their postnatal growth and neurodevelopment (Frank and Sosenko 1988; Hack, Breslau et al. 1991; Ehrenkranz, Younes et al. 1999; Hack, Schluchter et al. 2003; Cooke, Ainsworth et al. 2004; Hay 2008). The resultant increased morbidity and hospital stay increases the cost of treatment (Reddy, Deorari et al. 2000). The patho-physiology of feed intolerance in these high risk infants is poorly understood, limiting the therapeutic options. Delayed gastric emptying, intestinal immaturity, ileus of prematurity, and gastro-esophageal reflux may all play a role. It is not clear whether one or all of these mechanisms contribute to the observed feed intolerance. Thirty-seven per cent of infants born to mothers whose pregnancy was complicated by diabetes mellitus (IMDM) (Kitzmiller, Cloherty et al. 1978) also experience feed intolerance immediately after birth. This is in addition to the other well-known morbidities in this population of infants. These apparently healthy looking babies often require admission to the special care or transitional care unit until establishment of oral feeding which invariably leads to prolongation of parent-infant separation and hospital stay (Kitzmiller, Cloherty et al. 1978; Lee-Parritz 2004), and consequently cost of treatment. It is reasonable 22 Chapter 1 - Overview & Background to acknowledge that clinical practise has significantly changed in the last three decades for diabetic mothers and their infants and thus the actual incidence of feed intolerance or poor feeding may not be as high as previously reported. This may also explain the apparent absence of published literature on feed intolerance in IMDM. However with increasing incidence of diabetes in the general population, the clinical burden of IMDM and associated illnesses including feed intolerance may also increase. Therefore there is a need to understand the aetiology and pathophysiology of feed intolerance in this group of infants to formulate appropriate therapeutic interventions. Amylin is a 37 amino acid peptide hormone belonging to the calcitonin gene related peptide super family that is co-secreted with insulin from the pancreatic ß cells in response to enteral nutrient intake (Rink, Beaumont et al. 1993). In conjunction to playing a role in glucose homeostasis, it is recognised as a potent inhibitor of gastric emptying (Macdonald 1997). Amylin has been shown to delay gastric emptying in adult diabetic patients (Hoppener, Ahren et al. 2000) and critically ill children with feed intolerance (Mayer, Durward et al. 2002). There are no published data at the time of writing this dissertation on amylin and its role in the neonatal population. The focus of this dissertation is to establish whether amylin is responsible for feed intolerance in preterm infants and IMDM. 1.2 Study Rationale Amylin is a potent inhibitor of gastric emptying that may be linked to the pathophysiology of feed intolerance in the neonatal population. Understanding its role in new-born population may help develop therapeutic interventions which could lead to improved nutrition and outcomes. 1.3 Study Hypotheses It is hypothesized that serum amylin levels are raised in (i) infants of mothers whose pregnancy was complicated by diabetes and (ii) preterm infants with increased gastric residual volumes; and this is likely to be the principal cause of their feed intolerance. 23 Chapter 1 - Overview & Background 1.4 Study Aims The overall aims of this study were to determine serum amylin levels in (i) healthy preterm and term infants (control population), (ii) preterm and term IMDM and (iii) preterm infants with feed intolerance compared to preterm infants without feed intolerance. 1.5 Study Objectives 1.5.1 Primary Objectives The specific primary objectives of this study as detailed in (Table 1) were to determine serum amylin concentration (i) at birth and in the postnatal period in healthy preterm and term infants (ii) at birth and in the postnatal period in preterm and term IMDM and (iii) in preterm infants admitted to neonatal intensive care unit who subsequently experience feed intolerance compared to those who do not. 1.5.2 Secondary Objectives The specific secondary objectives of this study as detailed in (Table 1) were to investigate (i) the relationship between amylin and birth weight / gestational age (ii) if amylin and insulin were indeed co-secreted (iii) agreement of amylin levels in paired umbilical cord arterial and venous blood (iv) the relationship of amylin concentrations with gastric residual volumes (GRV), time to reach full enteral feeds and length of hospital stay. Rationale for secondary objective (iii): Umbilical venous sampling is technically easy compared to umbilical arterial sampling and was chosen as the ideal route for taking blood for this study. However experience of fellow researchers whose studies involved blood sampling from umbilical cords suggested that occasionally it was difficult to gain access to the umbilical vein necessitating umbilical arterial sampling. Secondly umbilical venous blood is thought to represent blood from the placenta whilst umbilical arterial blood to represent baby’s blood. Hence the study team decided to investigate amylin levels in paired samples wherever possible to investigate differences if any in amylin levels to inform the current and future studies employing umbilical cord blood. 24 Chapter 1 - Overview & Background Table 1. Study Objectives & Outcome Variables Primary Objective To Primary Outcome Variable determine serum amylin Amylin (median and interquartile concentrations range) concentration in pmol/L. 1. at birth and postnatal period in healthy preterm and term infants, 2. at birth and postnatal period in preterm and term IMDM, and 3. in preterm infants with and without feed intolerance. Secondary Objectives Secondary Outcome Variables To investigate the relationship between Correlation Coefficient serum amylin concentrations and gestational age and birth weight To determine the relationship between Correlation Coefficient amylin and insulin levels to confirm cosecretion To determine agreement between Correlation Coefficient and Bland paired umbilical arterial and venous Altman method comparison test amylin levels To investigate the relationship between Correlation Coefficient serum amylin concentrations and influential covariates (gastric residual volumes (GRV), time to full enteral feeds and length of hospital stay). 25 Chapter 2 – Literature Review: Amylin Peptide Chapter 2- Literature Review: Amylin Peptide 2.1 Introduction & Background Amylin or Islet amyloid polypeptide (IAPP) was first discovered in 1987 and is a second beta cell hormone that is co-located and co-secreted with insulin from pancreas (Figure 1). The names ‘Amylin’ and ‘islet amyloid polypeptide’ are often used interchangeably in current literature. The difference in nomenclature is largely geographical; European researchers tend to prefer IAPP whereas American researchers prefer Amylin. To maintain consistency ‘IAPP’ is referred to as ‘Amylin’ throughout this dissertation. Figure 1. Amylin is the second beta cell hormone This slide shows pictures (on the left) demonstrating that amylin and insulin are both present in the beta cells of islet of pancreas. A graphical representation of the structure of the amylin molecule is shown on the right. 2.2 Amylin Synthesis, Sources, Release & Metabolism Amylin is co-synthesized, co-packaged within the Golgi apparatus, and cosecreted within the secretory granule by the islet beta cells in response to 26 Chapter 2 – Literature Review: Amylin Peptide elevations of plasma glucose following enteral nutrient intake (Rink, Beaumont et al. 1993; Cooper 1994; Wimalawansa 1997). Although the major site of amylin biosynthesis lies in the pancreatic islet beta cells, animal studies have identified secondary sites in the endocrine cells of gastric mucosa (Mulder, Lindh et al. 1994; Mulder, Ekelund et al. 1997), dorsal root ganglias (Ferrier, Pierson et al. 1989; Fan, Westermark et al. 1994; Mulder, Leckstrom et al. 1995), and in the developing kidney (Wookey, Tikellis et al. 1998). Large number of amylin-immunoreactive cells were also found in the pyloric antrum, and a small number in the body of the stomach, duodenum and rectum in both man and rat (Toshimori, Narita et al. 1990). Amylin peptide may be prepared in vitro from diabetic pancreas by solubilisation of amyloid and isolation of the peptide by gel filtration and reverse phase chromatography. Amylin may also be produced by recombinant DNA techniques using the disclosed nucleic acid sequences. However one of the major problems with amylin is its spontaneous ability to deamidate and aggregate (Nilsson, Driscoll et al. 2002) rendering it insoluble and toxic for human administration. Amylin release into the circulation is increased postprandially and is in proportion to the amount of digested food (Bronsky, Chada et al. 2002). Oral and intravenous dextrose loads also promote amylin release possibly due to cosecretion with insulin in response to hyperglycemia. On the other hand, amylin release is inhibited by both hypoglycaemia and somatostatin (Mitsukawa, Takemura et al. 1990). Amylin is co-released with insulin in a molar ratio of approximately 1 to 100 and its diurnal profile corresponds to insulin in healthy non-diabetic subjects in response to carbohydrate and protein containing meals (Figure 2) (Butler, Chou et al. 1990; Mitsukawa, Takemura et al. 1990; Koda JE and JF 1995). 27 Chapter 2 – Literature Review: Amylin Peptide Figure 2. Twenty four hour plasma profile of amylin and insulin in healthy adults Twenty-four hour plasma profile of amylin is similar to insulin in healthy adults with low baseline levels that rapidly increase in association with increase in plasma glucose concentrations. Image courtesy of (Koda JE and JF 1995). In normal subjects plasma levels of amylin vary from fasting level of 5 pmol/L to 30 pmol/L postprandially (Bronsky, Chada et al. 2002). There is no difference in the mean (SD) amylin concentration in lean subjects (3.4 ± 0.7 pmol/L) and obese subjects (4.7 ± 0.9 pmol/L) with normal glucose tolerance (Hartter, Svoboda et al. 1991; Reda, Geliebter et al. 2002). In contrast amylin secretion following sustacal meal was observed to be absent in patients with type 1 diabetes and impaired markedly in patients with type 2 diabetes on insulin therapy (Figure 3) (Fineman MS 1996). 28 Chapter 2 – Literature Review: Amylin Peptide Figure 3. Amylin response after a liquid sustacal meal in patients with type 1 & type 2 diabetes mellitus The graph shows almost absence of amylin secretion in patients with type 1 diabetes, and its secretion is markedly impaired in insulin-requiring patients with type 2 diabetes. Image courtesy of (Fineman MS 1996). In healthy adults during glucose stimulation, like insulin, amylin concentrations exhibit high-frequency oscillatory pattern of nonglycosylated as well as glycosylated forms (Figure 4). The amylin frequency (min/pulse), burst mass (pM/burst), burst amplitude (pM/min), and basal secretion (pM/min) were 6.3 ± 1.0, 1.63 ± 0.88, 0.77 ± 0.4 and 0.04 ± 0.2. Whether basal amylin concentrations also exhibit an oscillatory pattern is not known (Juhl, Porksen et al. 2000). 29 Chapter 2 – Literature Review: Amylin Peptide Figure 4. Representative concentration profiles of amylin (A), total amylin immunoreactivity (TAI) (B) and insulin (C) This graph shows concentration profiles of amylin, total amylin immunoreactivity and insulin during constant glucose infusion (2 mg/ kg / min). Image courtesy of (Juhl, Porksen et al. 2000). The mean removal time of amylin in plasma is longer than insulin and similar to C-peptide (Clodi, Thomaseth et al. 1998), despite amylin having a faster clearance rate than insulin at the kidney (Thomaseth, Kautzky-Willer et al. 1996). 2.3 Amylin Structure, Gene & Relation to CGRP Superfamily Human amylin has 37 amino-acid sequences KCNTATCATQRLANFLVHSSNNFGAILSSTNVGSNTY, with a disulfide bridge between cysteine residues 2 and 7 and is characterised by a NH2-terminal cyclic ring structure and an amidated COOH-terminus (Figure 1, Figure 6 & Error! Reference source not found.). Both the amidated C terminus and the disulphide bridge are essential for the full biological activity of amylin (Cooper, 30 Chapter 2 – Literature Review: Amylin Peptide Day et al. 1989; Roberts, Leighton et al. 1989). The dynamic alpha helix structure of human amylin is shown in Figure 5. Figure 5. Alpha helix structure of human amylin Image courtesy of http://wiki.verkata.com/en/wiki/Amylin The human amylin gene is located on the short arm of chromosome 12 (Cooper, Day et al. 1989) and encodes the complete polypeptide precursor in two exons which are separated by an intron of approximately 5 kb transcribing an 89 amino acid precursor peptide (Nishi, Sanke et al. 1989). The proprotein convertase PC2 is primarily responsible for amylin processing within the secretory granule by converting the 89 aminoacid prohormone to the actively secreted 37 amino acid amylin (Badman, Shennan et al. 1996). Amylin belongs to the calcitonin gene related peptide superfamily (Rink, Beaumont et al. 1993) which in addition consists of calcitonin (CT), calcitonin gene-related peptide (CGRP) and adrenomedullin. CT and CGRP are derived from the CT/CGRP gene, which is encoded on chromosome 11. Chromosome 12 is thought to be an evolutionary duplication of chromosome 11 as amylin has 20% sequence homology with calcitonin and adrenomedullin and 44% 31 Chapter 2 – Literature Review: Amylin Peptide homology with calcitonin gene-related peptide (Bronsky, Chada et al. 2002). Importantly, a portion of the B-chain of insulin is strongly homologous to these four peptides. Thus amylin, CGRP, CT, and adrenomedullin are related to the insulin gene superfamily of peptides, which may all have diverged from a common ancestral gene during evolution and therefore may share some of the characteristics of insulin (Wimalawansa 1997). 2.4 Feto-Maternal Passage of Amylin Information with regards to transplacental passage of amylin is lacking at the time of writing this dissertation. The molecular weight of human amylin is 3904 Da compared to 5808 Da for human insulin. Published literature regarding transplacental passage of insulin may shed some light on passage of amylin across feto-maternal barrier. Early in vitro studies suggested that insulin does not cross the human placenta (Keller and Krohmer 1968; Sabata, Frerichs et al. 1970). However a subsequent placental perfusion study demonstrated that a small amount of human insulin, representing 1–5% of 59, 104, 448, and 1,198 μU/ml insulin concentration in the maternal artery (corresponding to subcutaneous doses of insulin of approximately 14, 24, 104, and 278 units respectively), was transferred from the maternal to the fetal circulation (Challier, Hauguel et al. 1986). Similarly maternal insulin lispro concentrations of ≥580 μU/ml (corresponding to subcutaneous dose of insulin lispro of approximately 75 units) led to a small transfer of insulin lispro across the placenta (Boskovic, Feig et al. 2003). In summary both human insulin and insulin lispro are unlikely to transfer across term placenta at clinically relevant therapeutic doses. Thus it may be assumed that endogenous amylin molecule has similar characteristic to insulin with regards to the transplacental passage. 2.5 Synthetic Analogue ‘Pramlintide’ Human amylin is insoluble and potentially toxic for human administration as a replacement in diabetic patients. Therefore synthetic, soluble, non-aggregating, equipotent (Fineman, Weyer et al. 2002; Symlin 2005) amylin analogue pramlintide acetate (Symlin, Amylin Pharmaceuticals) was developed by substitution of 3 residues with proline at positions 25 (alanine), 28 (serine), and 32 Chapter 2 – Literature Review: Amylin Peptide 29 (serine) as shown below in Figure 6. Pramlintide acetate (Symlin) is formulated as a clear, isotonic, sterile solution for subcutaneous administration. Figure 6. Comparison of aminoacid sequence of amylin and pramlintide Image is courtesy of http://diabetesmanager.pbworks.com Like amylin, pramlintide decreases post-prandial glucagon secretion, slows gastric emptying, and increases satiety. In addition, pramlintide results in weight loss, allows patients to use less insulin, lowers average blood sugar levels, and substantially reduces blood sugar surges that occurs in diabetic patients immediately after eating (Ratner, Want et al. 2002; Hollander, Levy et al. 2003; Riddle, Frias et al. 2007). The absolute bioavailability of a single subcutaneous dose of symlin is approximately 30 to 40% and approximately 40% of the drug is unbound in plasma and plasma concentrations peaks at 20 minutes, regardless of dose, and then declines over the subsequent 3 hours. It undergoes little or no hepatic metabolism and is cleared mainly via the kidneys, with a plasma half-life of 50 minutes (Weyer, Maggs et al. 2001). Pramlintide has been approved for use by the US Food and Drug Administration (FDA) in 2005 in patients with type 1 and type 2 diabetes who use insulin (Ryan, Jobe et 33 Chapter 2 – Literature Review: Amylin Peptide al. 2005). Hypoglycemia, nausea, vomiting, and anorexia were the most frequently reported adverse drug events associated with pramlintide therapy. Personal communication with Amylin Pharmaceuticals provided information regarding transplacental passage of pramlintide from studies performed on perfused human placenta. Term human placenta from uncomplicated pregnancy was obtained immediately after delivery. Pramlintide, at varying concentrations was introduced into the maternal reservoir. The maternal side of the placenta was perfused with constant concentration of pramlintide; the fetal circulation was closed. Samples were drawn from both the maternal and fetal circulations at regular intervals. The appearance of pramlintide in the fetal circulation was analyzed by a specific ELISA. The resulting data demonstrated that pramlintide had low potential (maternal to fetal ratio >1000/1) to cross the maternal-fetal placental barrier (Amylin Pharmaceuticals 2003). These results however cannot be extrapolated directly to the endogenous amylin and therefore in vivo and in vitro studies are required to investigate transplacental passage. 2.6 Amylin Receptors & Signal Transduction Mechanisms The calcitonin (CT) family of peptides utilize two seven transmembrane G protein-coupled receptors known as calcitonin receptor (CTR) and the calcitonin receptor-like receptor (CRLR). These receptors can dimerize with three different single transmembrane spanning proteins called Receptor activity-modifying protein (RAMP). When associated with a calcitonin receptor, RAMP alters its pharmacology from calcitonin-preferring to amylin-preferring. When coexpressed with RAMP1, RAMP2, or RAMP3 (Hay, Christopoulos et al. 2004), the CTR/RAMP complexes generate pharmacologically distinct amylin receptors, AMY1, AMY2, and AMY3, respectively (Poyner, Sexton et al. 2002) which bind with high affinity to amylin. The binding sites at nucleus accumbens and subfornical organ are composed of CTR/RAMP1 (AMY1) whereas the binding sites at area postrema are composed of CTR/RAMP3 (AMY3) (Young 2005). Despite pharmacological evidence of amylin sensitivity in several peripheral tissues, selective amylin binding outside of the brain is observed only in the renal cortex. 34 Chapter 2 – Literature Review: Amylin Peptide 2.7 Amylin Receptor Antagonist Peptides such as AC187, salmon calcitonin (sCT), AC66, CGRP [8-37], which are typically analogues of truncated salmon calcitonin, have been developed as potent and selective amylin antagonists and have been useful in identifying amylinergic responses. Antagonism of a response with an order of potency of AC187> AC66 > CGRP [8-37] is suggestive that it is mediated via amylin receptors. Activation of a response with salmon calcitonin (sCT) > amylin > calcitonin gene-related peptide (CGRP) > mammalian CT suggests activation via the AMY1 receptor, while sCT = amylin >> CGRP > mammalian CT suggests activation via AMY3 receptors (Young A, 2005). AC-187 is particularly selective and potent at blocking amylin receptors (Young, Gedulin et al. 1994) and most often cited in studies using amylin antagonists. It also blocks amylin mediated excitatory effect on area postrema neurons and subfornical organ neurons. In addition it blocks amylin induced c-Fos expression in the area postrema (Riediger, Schmid et al. 2001). 2.8 Biological Functions of Amylin Amylin has a number of biological functions both in animals and humans of which inhibition of post prandial glucagon secretion, delaying gastric emptying, and reduction of appetite and food intake are its most potent role. Amylin plays a vital role in maintaining glucose homeostasis by virtue of its activity on the stomach and glucagon secretion which is illustrated in Figure 7. 35 Chapter 2 – Literature Review: Amylin Peptide Figure 7. Role of amylin in regulation of postprandial glycemia This slide demonstrates the actions of amylin in regulation of blood glucose in fed state. Following ingestion of food the rise in blood glucose stimulates the pancreas to secrete insulin along with amylin which proceeds through the circulation to reach the brain where it binds to nuclei to slow gastric emptying and decrease food intake both of which delay the delivery of nutrients to the circulation. In addition amylin reduces hyperglucagonemia in the postprandial period, thereby reducing the delivery of glucose from the liver into the circulation. Thus blood glucose is maintained by fine tuning of the rate of glucose appearance and glucose disappearance in the blood. Following ingestion of nutrient intake, glucose is released into the blood stream which stimulates the co-secretion of insulin and amylin from pancreatic beta cells. Amylin regulates postprandial glycaemia by suppression of glucagon release (Ludvik, Thomaseth et al. 2003), delaying gastric emptying (Kolterman, Gottlieb et al. 1995; Young, Gedulin et al. 1995; Young 2005; Young 2005), inhibition of glucose release and hepatic glucose production (Ludvik, KautzkyWiller et al. 1997) and possibly by noncompetitively antagonizing the effect of 36 Chapter 2 – Literature Review: Amylin Peptide insulin and increasing the activity of the glycogen synthase kinase 3 (GSK 3) (Abaffy and Cooper 2004). The most reported role in literature of amylin and pramlintide is inhibition of gastric emptying in animals (Young, Gedulin et al. 1995; Clementi, Caruso et al. 1996; Young, Gedulin et al. 1996), adult patients with type 1 and type 2 diabetes (Kong, King et al. 1997; Kong, Stubbs et al. 1998; Vella, Lee et al. 2002) and non-diabetic subjects (Samsom, Szarka et al. 2000). The effect of amylin on gastric emptying is greater than that produced by other gut peptides, such as gastric inhibitory peptide (GIP), cholecystokinin (CCK), and glucagon like peptide (GLP-1) (Young, Gedulin et al. 1996). In particular, amylin is 15- to 20 fold more potent than GLP-1 and CCK (Gedulin 1996; Young, Gedulin et al. 1996). It is also known to inhibit gastric acid secretion and maintain gastric mucosal integrity (Young 2005). Amylin is also known to control the central mechanisms responsible for food intake and for the feeling of satiety and thirst. Amylin decreases food intake (Morley and Flood 1991; Lutz, Del Prete et al. 1994; Lutz, Geary et al. 1995; Bronsky, Chada et al. 2002) mainly through reduction in meal size (Lutz, Geary et al. 1995; Mollet, Gilg et al. 2004) and increasing satiety (Morley and Flood 1991; Lutz, Del Prete et al. 1994). Amylin appears to reduce food intake centrally by activation of receptors in area postrema, a circumventricular organ outside the blood brain barrier involved in regulating food intake and peripherally through augmenting the actions of other gut peptides such as CCK, glucagon, and bombesin, all of which inhibit food intake by stimulating ascending vagal fibers and secretion of amylin. In addition amylin may decrease food intake by inducing anorexia through its effect on brain by increasing the transport of the precursor tryptophan into the brain to inhibit feeding and increase satiation by serotonin action in the paraventricular nucleus, stimulating histamine H1 receptors, inhibition of stimulation of feeding by potent hypothalamic neuropeptide Y (NPY) and peripherally by inhibition of nitric oxide which is a major regulatory agent in the gastrointestinal tract (Reda, Geliebter et al. 2002). Amylin significantly increased water intake in euhydrated rats by activating neurons in the subfornical organ of the brain suggesting amylin release during food intake may stimulate prandial drinking (Riediger, Rauch et al. 1999). 37 Chapter 2 – Literature Review: Amylin Peptide Amylin has also been found to be a growth factor for cells involved in bone metabolism (Cornish, Callon et al. 1995; Villa, Rubinacci et al. 1997; Cornish, Callon et al. 2001), particularly for the proliferation of osteoblasts (Cornish, Callon et al. 1995; Cornish, Callon et al. 1998) and differentiation of osteoclast (Cornish, Callon et al. 2001) and inhibition of bone resorption (Bronsky, Chada et al. 2002) which helps increase bone density and bone mass (Cornish and Naot 2002). Amylin is found to be a growth factor for isolated fetal beta cells which may have a role in the development of fetal pancreas (Karlsson and Sandler 2001). Amylin is a potent proliferation factor both during the development of proximal tubules (Wookey, Tikellis et al. 1998) and in the proliferation of adult epithelial cells (Harris, Cooper et al. 1997). Amylin at physiological doses has been shown to stimulate renin activity in both rats (Wookey, Tikellis et al. 1996) and man (Cooper, McNally et al. 1995) and demonstrates a modest pressor effect (Cooper, McNally et al. 1995; Haynes, Hodgson et al. 1997) possibly mediated by insulin-induced changes in rennin release (Ikeda, Iwata et al. 2001). In addition renal amylin binding sites in proximal tubules could be correlated with blood pressure in rat models of renal hypertension (Cao, Wookey et al. 1997; Wookey, Cao et al. 1997; Wookey, Cao et al. 1998; Wookey and Cooper 1998). Amylin has been reported to induce relaxation of aortic rings (Luk'yantseva, Sergeev et al. 2001), inhibit CCK induced contraction of smooth muscle cells (Ochiai, Chijiiwa et al. 2001), and decrease pulmonary vascular tone (Golpon, Puechner et al. 2001). Amylin modulates insulin sensitivity of skeletal muscle (Bronsky, Chada et al. 2002). Amylin is vasoactive and produces hypotension and vasodilatation (Young, Rink et al. 1993; Bronsky, Chada et al. 2002) probably by inhibiting the secretion of the atrial natriuretic peptide through CGRP1 and CT receptors (Piao, Cao et al. 2004), and exerts anti-inflammatory activity (Hall and Brain 1999) possibly due to cross reactions with CGRP receptors (Zhu, Dudley et al. 1991). Like CGRP amylin may also play a role in maternal sepsis and shock (Parida, Schneider et al. 1998). 38 Chapter 2 – Literature Review: Amylin Peptide 2.9 Mechanisms of Amylin Action at Molecular level Experimental investigations in rodents indicate that the gastrointestinal effects of amylin are mediated via a central pathway that involves the area postrema and visceral efferents of the vagus nerve. The area postrema in the brainstem which contains a high density of amylin binding sites (Figure 7) is exposed to changes in plasma amylin and specifically co-excited by glucose (Riediger, Schmid et al. 2002) because it does not have a blood-brain barrier (Gross 1992). As there is no unique amylin receptor gene, the most likely mechanism of amylin action at molecular level in the brain and elsewhere is likely to be through CTR and RAMPs bestowing functionality to the amylin receptors and binding sites (Figure 8). Figure 8. Amylin binding sites in rat brain An audioradiograph of the rat brain demonstrating major binding sites containing amylin receptors in nucleus accumbens, the dorsal raphe, and the area postrema which are thought to be important in regulation of appetite as well as sympathetic and parasympathetic tone. 39 Chapter 2 – Literature Review: Amylin Peptide In addition peripherally injected amylin strongly activates neurons of nucleus tractus solitarius (Rowland and Richmond 1999; Riediger, Schmid et al. 2002) which is neuronally interconnected with the area postrema (van der Kooy and Koda 1983; Shapiro and Miselis 1985). Selective lesioning of the area postrema and/or bilateral vagotomy abolishes the effect of amylin on gastric emptying (Jodka 1996; Edwards 1998) demonstrating the importance of this central pathway in mediating amylin’s physiological functions. In addition the effect of amylin to slow gastric emptying is overridden in the presence of hypoglycaemia (Gedulin and Young 1998). Amylin exerts its inhibitory action on glucagon by mechanisms extrinsic to pancreas (Silvestre, Rodriguez-Gallardo et al. 2001). Thus amylin contributes to metabolic control by regulating nutrient influx into the blood resulting in increased glucose disposal and slowing nutrient delivery and postprandial hyperglycemia. Two main regions of the circumventricular organs in the rat brain (Figure 9) namely subfornical organ and area postrema mediate peripheral variations in amylin levels via receptors including drinking (Schmid, Rauch et al. 1998; Riediger, Rauch et al. 1999; Riediger, Schmid et al. 1999) and feeding behaviour (Lutz, Senn et al. 1998; Riediger, Schmid et al. 2001) respectively. 40 Chapter 2 – Literature Review: Amylin Peptide Figure 9. Location of amylin action within circumventricular organs in rat brain The circumventricular region contains a rich capillary plexus and has a fenestrated endothelium rendering the structures outside of the blood brain barrier. It is made up of 7 organs namely median eminence, organum vasculosum of the lamina terminalis, subfornical organ, choroid plexus, pineal gland, and subcommissural organ and area postrema. Image courtesy of http://www.endotext.org Additionally peripherally applied amylin inhibits feeding through binding sites in area postrema and nucleus accumbens (Sexton, Paxinos et al. 1994; van Rossum, Menard et al. 1994; Lutz, Senn et al. 1998; Lutz, Mollet et al. 2001; Bronsky, Chada et al. 2002) which may be processing these peripheral signals (Baldo and Kelley 2001) and is independent of vagal afferents (Lutz, Del Prete et al. 1995). Amylin may induce anorexia through stimulating H1 receptors (Mollet, Lutz et al. 2001) and this effect is attenuated with dopamine D2 receptor antagonists (Lutz, Tschudy et al. 2001). Amylin also inhibits stimulation of feeding centrally by the potent hypothalamic neuropeptide Y (NPY) (Morris and Nguyen 2001) and peripherally by inhibiting nitric oxide (Morley and Flood 1994; Morley, Flood et al. 1995). 41 Chapter 2 – Literature Review: Amylin Peptide In summary amylin is a neuroendocrine hormone which helps in regulating feeding behaviour by reducing meal size, increasing satiety and anorexia by stimulating neurons particularly in the area postrema and hindbrain. 2.10 History of Discovery of Amylin Although the presence of amyloid deposits within the islet was first described at the beginning of the 20th century, it was not until 1987 that the structure of amylin was discovered (Cooper, Willis et al. 1987; Westermark, Wernstedt et al. 1987; Cooper, Leighton et al. 1988). The history leading to the discovery of amylin is an interesting story and began in 1869 when Paul Langerhans (Langerhans 1869; Morrison 1937) described the existence of acinar glandular cells and pairs or groups of small polygonal cells with round nuclei in the interacinar spaces of rabbit’s pancreas. Oscar Minkowski in 1889 observed the connection between pancreatic secretion and diabetes on depancreatized dogs (Bliss 1982). It was Languesse E (Languesse 1893) in 1893 who named the clumps of cells observed by Paul Langerhans as the ‘Islands or Islets of Langerhans’. Eugene Opie in 1889 was the first to describe "hyaline degeneration of the islands of Langerhans" in the pancreas of patients with hyperglycemia and in 1901 provided its pathological link to diabetes (Opie 1901). It was not until 1922 however, that a Canadian team comprising of Banting, Best, Macleod and Collip discovered insulin (Banting 1922). Thirty-three years later, in 1955, the protein structure of insulin was completely sequenced by Frederick Sanger (Ryle, Sanger et al. 1955). The amyloidal nature of the hyaline material described by Eugene Opie was established by Ahronheim in 1943 and later confirmed by Ehrlick and Ratner in 1961 (Ahronheim 1943; Ehrlich and Ratner 1961). Westermark in 1973 demonstrated the fibrillar structure (Westermark 1973) of the hyaline staining material on electron microscopy. Importantly, in 1987 he also discovered that it was predominantly made up of a 37 amino acid monomer and referred to it as “islet amyloid polypeptide” (IAPP). Interestingly in the same year Cooper from another laboratory also discovered this 37 amino acid polypeptide in pancreatic amyloid deposits and named it “amylin” (Cooper, Willis et al. 1987; Westermark, Wernstedt et al. 1987; Cooper, Leighton et al. 1988). 42 Chapter 2 – Literature Review: Amylin Peptide 2.11 Association of Amylin with Islet Amyloid Deposits in Diabetes Amylin is associated with type 2 diabetes, a disease that has increased over the past two decades and now afflicts an estimated 100 million worldwide (Hossain, Kawar et al. 2007). At post-mortem 95% patients with type 2 diabetes stain positive for amyloid deposits in the pancreas composed of mature, fibrillar amylin (Hoppener, Ahren et al. 2000). Since 1987 there has been a rapid increase in publications on amylin most probably due to the fact that islet amyloid in type 2 diabetes mellitus is strongly associated with islet beta cell loss (Hayden and Tyagi 2001; Jaikaran and Clark 2001). Additionally, it is also acknowledged that amylin may be linked to the pathogenesis and therefore a potential target for the treatment of diabetes (Schmitz, Brock et al. 2004). 2.12 Amyloidosis and Composition of Amyloid Deposits Amyloidosis refers to group of disorders characterised by abnormal deposition of amyloid proteins in organs and/or tissues. The clinical classification of amyloidosis includes primary (de novo), secondary (a complication of a previously existing disorder), familial, and isolated types. Out of the approximately 60 amyloid proteins which have been identified so far (Mok, Pettersson et al. 2007), at least 36 have been associated in some way with a human disease (Pettersson-Kastberg, Aits et al. 2008) such as Bovine spongiform encephalopathy (prion protein PrPSc), Alzheimer’s disease (betaamyloid), Parkinson’s (Alpha-synuclein) and Creutzfeldt-Jakob disease (prion protein PrPSc). A characteristic feature of amyloid deposit histologically is the positive staining with Congo red (Figure 10) (Satoskar, Burdge et al. 2007) and birefringence on viewing with polarised light (Figure 11) (Hayden and Tyagi 2001). 43 Chapter 2 – Literature Review: Amylin Peptide Figure 10. Amyloid deposit in pancreatic islet High power microscopic view showing a pancreatic islet with extensive deposition of a pink homogenous material (Congo red stain) that has features suggestive of amyloid surrounded by normal pancreatic acini. Image courtesy of http://pathcuric1.swmed.edu/PathDem0/end6/end660.htm 44 Chapter 2 – Literature Review: Amylin Peptide Figure 11. Amyloid deposit in lymph node showing birefringence Congo red staining of amyloid deposit in the lymph node demonstrating green birefringence to polarised light. Image courtesy of http://www.flickr.com/photos/euthman/377559955/ Deposition patterns of amyloid vary between patients but each disease is characterized by a particular protein or polypeptide that aggregates in an ordered manner to form insoluble amyloid fibrils that are deposited in the tissues, where they have been associated with the pathology of the disease. These different proteins and peptides, are characterised by common structural features, whereby stacks of parallel peptide strands (Jayasinghe and Langen 2004) run perpendicular to the fibril axis hydrogen-bond (Sumner Makin and Serpell 2004) to form extended beta-sheets (Error! Reference source not found.). 45 Chapter 2 – Literature Review: Amylin Peptide Figure 12. Extended cross-beta structure of amyloid Extended cross-beta sheet amyloid structure consisting of layers of aminoacid chains lying perpendicular to the fiber axis. Image courtesy of http://www.vanderbilt.edu/vicb/ Similarly one of the major polypeptide components forming the monomeric unit of polymerized, aggregated, and beta pleated sheet structure of islet amyloid in type 2 diabetic patients is amylin (Figure 13) (Hoppener, Ahren et al. 2000; Kajava, Aebi et al. 2005). 46 Chapter 2 – Literature Review: Amylin Peptide Figure 13. Superpleated beta structure of amyloid fibrils The superpleated beta-structural model of amyloid fibrils of human amylin viewed along the fibril axis. Image courtesy of (Kajava, Aebi et al. 2005) Amylin is implicated in the pathogenesis of type 2 diabetes and Alzheimer’s due to its ability to form amyloid deposits leading to destruction of cell membranes (Green, Goldsbury et al. 2004; Sedman, Allen et al. 2005). Amylin receptors may play an important role in the neurotoxic effects of amyloid beta and therefore its inhibition could be promising in the treatment of Alzheimer’s disease (Kurganov, Doh et al. 2004) Sequences of human proamylin that may contribute to the processes of amyloid formation include phenylalanine-23 (Azriel and Gazit 2001) and amino and carboxy terminal regions (Jaikaran and Clark 2001; Jaikaran, Higham et al. 2001; Padrick and Miranker 2001). Amylin and its 10-residue fragment between amino acids 20 and 29 (Error! Reference source not found.) is known to spontaneously deamidate, and the presence of less than 5% of deamidation impurities leads to the formation of aggregates by the purified peptide that has the hallmarks of amyloid (Nilsson, Driscoll et al. 2002). 47 Chapter 2 – Literature Review: Amylin Peptide Figure 14. Region responsible for amyloid formation The primary structure of amylin showing region responsible for amyloid formation. Image courtesy of http://www.joplink.net/prev/200507/02.html 2.13 Laboratory Assays Studies investigating the biological roles of amylin in health and disease require sensitive and specific assays capable of measuring amylin at the concentrations at which it circulates in plasma. Some of the Radioimmunoassay’s (RIAs) developed initially to measure human amylin were reported to be sensitive enough to detect fasting concentration of amylin. However RIA is labour intensive, time consuming taking up to 5 days, extraction and assay procedure could potentially introduce variability into the results and required up to 5 ml blood volume to obtain enough plasma before assay to extract amylin. This blood volume would be unsafe and thus prohibitive especially in preterm and very low birth weight population. To combat the problems highlighted with RIA, two sandwich-type immunoassays were developed which are considered to be rapid, sensitive, analyze large numbers of samples within a few hours and capable of measuring 0.5 to 2 pmol/L amylin in 50 µL of plasma. Furthermore, 48 Chapter 2 – Literature Review: Amylin Peptide these two assays exhibited limited cross reactivity (< 0.01 %) with other related peptides, detectable dynamic range of amylin concentration 2 to 100 pmol/L, average intraassay coefficient of variations (CVs) of <10%, average interassay CVs of <15%, and good linearity on dilution and recovery of added amylin. Thus the availability of these assays made it possible to study plasma amylin concentrations in various disease states in different age groups including neonatal population. 49 Chapter 3 – Literature Review: Gastric Emptying Chapter 3- Literature Review: Gastric Emptying 3.1 Anatomy and Physiology of Gastric Motor Function An understanding of the pathogenesis of delayed gastric emptying may be helped by revisiting the anatomy and physiology of normal gastric motor function. The stomach is divided into cardia, fundus, body and pylorus each with different cells and functions. The oesophageal and pyloric sphincters help to keep the contents of the stomach contained. In adult humans, the anatomic capacity of stomach is about 45 ml which normally expands to hold about 1-3 litres of food. The stomach volume in the fetus is approximately 10 ml which increases to 1821 ml by term (Nagata, Koyanagi et al. 1994; Sase, Miwa et al. 2005). The anatomic capacity of new-born stomach depends on birth weight and varies from 22-38 ml in 1.5-4.0+ kg infants which increases to 45 ml in 1 week and 75 ml in 2 weeks postnatal age. The physiologic capacity is less than the anatomic capacity at birth, equals by day 5 and reaches 81 ml by day 10 in 2.0-4.0+ kg infants (Scammon RE 1920). The gastric motor function is controlled by parasympathetic and sympathetic nervous systems, enteric brain neurons and smooth muscle cells. Extrinsic neural controls by parasympathetic pathways are conveyed to the stomach and upper intestine through the vagal efferents arising in the dorsal motor nucleus of the vagus nerve, nucleus ambiguus and tractus solitaries which form characteristic bead chain like terminals in the myenteric plexus throughout the stomach and upper intestine but without directly innervating muscle (Berthoug 1993). The vagus nerve regulates the distribution of food between proximal and distal regions of the stomach. The proximal stomach serves as a reservoir for solid foods and induces tonic pressure to facilitate liquid emptying. At the distal end of the stomach, the tone of the pylorus controls the rate at which the contents of the stomach are emptied into the small intestine. During fasting state both adults as well as term infants exhibit migrating motor complexes (MMCs) which are peristaltic waves originating in the stomach that progress through small intestine into colon approximately every 75-90 minutes and are interrupted by a meal. As the distal stomach grinds ingested solid foods, phasic 50 Chapter 3 – Literature Review: Gastric Emptying and tonic pyloric pressures in conjunction with duodenal contractions slow gastric emptying, which in turn disrupts MMC to allow for normal digestion and absorption of nutrients (Chaikomin, Rayner et al. 2006). In summary, normal gastrointestinal motor function is a complex sequence of events that is controlled by an extrinsic nerve supply from the brain and spinal cord, the complex plexi within the wall of the stomach and intestine, and the effects of locally released amines and peptides that alter the excitability of the smooth muscle of the intestine. Abnormalities in any of these locations can lead to delayed gastric emptying. 3.2 Regulation of Gastric Emptying Carbohydrate absorption is crucial to understanding the control of gastric emptying. In the normal adult human, a moderately sized meal contains up to 50-100 g of glucose but the total amount of glucose in plasma is approximately 5 g (Young, Pittner et al. 1995). Thus without regulation of its absorption, glucose could easily exceed the normal level found in plasma. The glucose released into the blood stream following ingestion of food stimulates rapid secretion of insulin and simultaneous suppression of glucagon secretion. These reciprocal changes in islet hormones stimulate glucose uptake and suppress hepatic glucose production. At the same time, glucose transport and disposal are facilitated in peripheral tissues, mainly in muscle, and to some extent in kidney and adipose tissue (Kelley, Mitrakou et al. 1988; Meyer, Dostou et al. 2002). As blood glucose concentrations rise, the decelerated rate of gastric emptying contributes to maintenance of glucose homeostasis (Barnett and Owyang 1988). Furthermore, in the presence of acute hyperglycaemia, proximal gastric tone is diminished, frequency and propagation of antral pressure waves are suppressed, and pyloric contractions are stimulated, all of which contribute to the slowing of gastric emptying (Meyer, Dostou et al. 2002) as a protective mechanism to control glucose efflux into the blood stream. Hence there seems to be matching of fluxes via ‘entero-insular loop’ to ensure regulated carbohydrate efflux from the stomach for absorption is approximately equal to the regulated insulin-mediated uptake from the plasma into peripheral tissue. This regulatory system which is essential for normal control of blood glucose is disrupted in amylin-deficient state such as type-1 diabetes. 51 Chapter 3 – Literature Review: Gastric Emptying The rate of gastric emptying in adults is also influenced by a number of other factors, including the size, composition (fat versus carbohydrate) and type (liquid versus solid) of the meal (Moore, Christian et al. 1981; Collins, Horowitz et al. 1983), and activity of the gut hormones (Macdonald 1997). Osmolarity and temperature of feed, posture, and stress also impact the gastric emptying rate (Hunt and Knox 1968; Blair, Wing et al. 1991). Small meals empty more rapidly than large ones and a meal with a high fat content has a higher half time for gastric emptying than a meal with high carbohydrate content (Sidery, Macdonald et al. 1994). The rate of gastric emptying of liquids is faster than that of solids (Collins, Houghton et al. 1991), and addition of soluble fiber to food delays gastric emptying rate (Torsdottir, Alpsten et al. 1991; Frost, Brynes et al. 2003). 3.3 Role of Other Emptying Gastrointestinal Peptides on Gastric Gastric emptying is modulated by feedback mechanisms arising from the interaction of nutrients from food with the small intestine (Chaikomin, Rayner et al. 2006) and intestinal peptides including glucagon-like peptide 1 (GLP-1), CCK, secretin, peptide YY, gastrin-releasing peptide (bombesin), and amylin. GLP-1 inhibits gastric emptying; its secretion stimulated by the intestinal nutrient content and flow rather than by the plasma glucose concentration itself (Schirra, Katschinski et al. 1996; Schirra, Leicht et al. 1998). On the other hand, amylin inhibits gastric emptying secondary to release of insulin due to postpandrial hyperglycemia with amylin concentrations increasing from 9 to 43 pmol/L (Samsom, Szarka et al. 2000; Woerle, Albrecht et al. 2008). Of the reported peptides known to inhibit gastric emptying, amylin is the most potent of the endogenous peptides identified to date (Young, Gedulin et al. 1996). In contrast to amylin, Ghrelin is synthesised predominantly by the stomach and is the endogenous ligand for the growth hormone secretagogue receptor which is expressed in brain stem and hypothalamic nuclei (Kojima, Hosoda et al. 1999). Exogenous ghrelin causes large meals to be eaten (Tschop, Smiley et al. 2000; Wren, Seal et al. 2001; Wren, Small et al. 2001) and is a most potent stimulator of gastric contractions and emptying (Hellstrom, Gryback et al. 2006). Other gut hormones such as gastrin, PP, CCK, secretin, GLP-1, GLP-2, oxyntomodulin, 52 Chapter 3 – Literature Review: Gastric Emptying and PYY also influence gastrointestinal motility and may potentiate amylin’s inhibition of gastric emptying (Figure 15). Figure 15. Site of synthesis of main gut hormones influencing appetite and gastric emptying These gut hormones may also potentiate amylin’s role in delaying gastric emptying. CCK, cholecystokinin; PYY, peptide YY; GLP1, glucagon-like peptide 1; GLP2, glucagon-like peptide 2. Image courtesy of (Druce and Bloom 2006) 3.4 Diabetes and its Effects on Gastric Emptying Diabetes is frequently associated with symptoms due to disturbances of gastrointestinal motility and/or sensation (Samsom, Roelofs et al. 1998; Bytzer, Talley et al. 2001). Approximately 30%-50% of patients with long standing type 1 and type 2 diabetes show delayed gastric emptying (Horowitz, O'Donovan et al. 2002; Samsom, Vermeijden et al. 2003) which may coexist with gastroparesis due to diabetic autonomic neuropathy (Samsom, Akkermans et al. 1997; Rayner, Samsom et al. 2001). 53 Chapter 3 – Literature Review: Gastric Emptying Type 1 diabetes patients have delayed gastric emptying rates in response to hyperglycemia (Schvarcz, Palmer et al. 1997) including patients with autonomic neuropathy probably due to changes in antroduodenal motility (Samsom, Akkermans et al. 1997). On the contrary, gastric emptying rate is accelerated in type 1 diabetics in insulin induced hypoglycaemia (Schvarcz, Palmer et al. 1993). Thus there is an inverse relationship between the rate of gastric emptying and the blood glucose concentration, so that emptying is slower during hyperglycaemia and faster during hypoglycaemia. Delayed gastric emptying also occurs frequently in type 2 diabetes mellitus partly due to increased plasma glucose concentrations (Ziegler, Schadewaldt et al. 1996) and partly due to raised amylin levels. However unchanged (Kong, King et al. 1999) or accelerated (Schwartz, Green et al. 1996) gastric emptying rates have also been reported. Although a review reports increase in amylin level in gestational diabetes (Ludvik, Kautzky-Willer et al. 1997), literature on the effects of gestational diabetes on gastric emptying is lacking. Because amylin is co-secreted by β-cells, individuals with diabetes have both insulin and amylin deficiency. Amylin deficiency is absolute in type 1 diabetics, whereas in type 2 diabetics, amylin levels increase in the early stage due to insulin resistance but decrease progressively in tandem with the decline in insulin secretion (Edelman and Weyer 2002). However contrary to the expectation of accelerated gastric emptying, even patients with type 1 diabetes demonstrate delayed gastric emptying suggesting factors other than amylin play a role in control of gastric motility (Heptulla, Rodriguez et al. 2008) in diabetic patients. Delayed gastric emptying in type 2 diabetes patients (Hoppener, Ahren et al. 2000) may be partly explained by raised amylin levels due to insulin resistance syndrome. Amylin’s synthetic analogue pramlintide slows liquid and solid gastric emptying by 1–2 hours through vagal inhibition in both type 1 and type 2 diabetic patients (Whitehouse, Kruger et al. 2002; Kellmeyer, Kesty et al. 2007). Thus gastric emptying in diabetes is influenced particularly by glycaemia with reciprocal relationship between the rate of gastric emptying and the blood 54 Chapter 3 – Literature Review: Gastric Emptying glucose concentration. Factors other than and partly due to amylin seem to be responsible for the gastrointestinal symptoms associated with delayed gastric emptying in patients with type 1and type 2 diabetes respectively. 3.5 Methods of Measurement of Gastric Emptying in Humans A variety of different techniques are available to study the rate of gastric emptying in humans. The majority of these tests are only applicable to adult patients. However methodological details and review of individual tests may help to establish relevance of these tests to the neonatal and paediatric setting. Early methods employed to measure gastric emptying such as dye dilution and single and double sampling gastric aspiration techniques (George 1968) are now almost obsolete due to the availability of more advanced techniques. Gastric emptying scintigraphy is considered gold standard as it is a physiologic, quantitative, non-invasive test for evaluating gastric emptying for both solids and liquids. It is validated in healthy subjects and also allows for assessment of intragastric distribution (Tougas, Chen et al. 2000; Abell, Camilleri et al. 2008; Abell, Camilleri et al. 2008). A radiolabelled 99mTc-sulphur colloid or 113mIn DTPA or 67Ga EDTA meal is ingested under standardized ambient noise, ambient light, and patient comfort. Imaging is performed while the patient is positioned constant either standing or sitting. The passage of the meal from the stomach to the small intestine is then followed by taking images at regular intervals with a gamma camera (Macdonald 1997). The main disadvantages of this procedure are its cost and exposure to a small amount of radiation which would be a limitation for its use in neonates’ especially preterm infants. The basis of the bio impedance technique (Savino, Cresi et al. 2004) is that after the ingestion of fluids with a different conductivity from body tissues, the impedance to an electrical current through the upper abdomen changes, impedance increases as the stomach fills and decreases as it empties. The slope of the plot of impedance versus time is used to calculate the time for emptying half of the meal giving a value for impedance half-life. Epigastric impedance patterns closely correlated with scintigraphy in the measurement of gastric emptying in infants (Savino, Cresi et al. 2004). 55 Chapter 3 – Literature Review: Gastric Emptying The 13C-octanoate breath test is a non-invasive approach for measuring gastric emptying, which correlates well with scintigraphy (Ziegler, Schadewaldt et al. 1996). When octanoic acid (with solid meal) or acetate (with liquid meal) labelled with 13C is combined with different food substrates, these compounds pass unabsorbed through the stomach to the duodenum but are quickly absorbed in the small intestine and metabolized to 13CO2 in the liver. Appearance of 13CO2 in the breath marks gastric emptying, absorption and excretion of 13CO2. This methodology offers significant advantages over scintigraphy and other methods in terms of cost, accessibility, and avoidance of radioactive materials. Sensitivity and specificity of the 13C-octanoate breath test for measuring gastric emptying has been shown to be 75% and 86%, respectively (Ziegler, Schadewaldt et al. 1996). In addition, because of low intraindividual variability, breath tests may be best applied to tracking gastric emptying during therapeutic interventions. It is noteworthy that the 13Coctanoate exhalation times correlate with gastric emptying but do not represent real gastric emptying times and the results may depend on intact liver and pulmonary function. 13C-Spirulina platensis breath test (Vella, Lee et al. 2002) is a slight variation to 13C-octanoate breath test, has recently been validated and deemed as reproducible as scintigraphy (Szarka, Camilleri et al. 2008). Ultrasonography (Holt, Cervantes et al. 1986) allows real-time changes in gastric motility and gastric emptying including transpyloric flow of liquid gastric contents, and accommodation in the proximal stomach (Berstad, Hausken et al. 1996; Kim, Myung et al. 2000; Parkman, Hasler et al. 2004). Ultrasound measurements of gastric emptying is well validated in healthy subjects and showed a high correlation to scintigraphy in quantifying emptying including liquids of both low and high nutrients (Benini, Sembenini et al. 1999). Although ultrasound methodologies are relatively low cost and will not expose patients to radioactive materials, their performance is operator-dependent and these studies are reliable only for measurement of liquid emptying rates (Parkman, Hasler et al. 2004). Also, it does not permit determination of the phase of the meal that is being emptied from the stomach. Recently, 3D ultrasonography has been shown to provide a valid measure of gastric emptying of liquid meals in healthy subjects (Gentilcore, Hausken et al. 2006). Real time ultrasound measurement of gastric antral cross sectional area (ACSA) was shown to be 56 Chapter 3 – Literature Review: Gastric Emptying reproducible in the assessment of gastric emptying in preterm infants (Ewer, Durbin et al. 1994). The technique is based on serial measurements of ACSA as it fills and empties during and after a feed. Ultrasound and scintigraphic techniques showed 90% concordance in subjects varying from 15 day infants to 10 year old children. The 10% discordant results were related to overlapping of duodenum and stomach during scintigraphy and shadowing of the gastric antrum by air during ultrasonography (Gomes, Hornoy et al. 2003). Magnetic resonance imaging is another non-invasive test that allows for measurement of gastric motility and emptying and can differentiate among an ingested meal, solid and liquid phases, gastric secretion, and air (Schwizer, Maecke et al. 1992; Feinle, Kunz et al. 1999). This technique strongly correlates with gastric emptying profiles obtained by scintigraphy for liquid and solid meals (Schwizer, Maecke et al. 1992; Feinle, Kunz et al. 1999). Although magnetic resonance imaging requires no exposure to radioactive materials, the procedure cost is high and it requires significant time for interpretation of test results (Parkman, Hasler et al. 2004). The appearance of paracetamol in the systemic circulation is an indirect method of determining the rate of gastric emptying. Because paracetomol after oral administration is not absorbed from the stomach but is rapidly absorbed from the small intestine, the area under the serum concentration curve (AUC) is determined by the rate of gastric emptying (Heading, Nimmo et al. 1973). Paracetamol absorption test (van Wyk, Sommers et al. 1995) is simple, reproducible, and accurate bedside tool (Sanaka, Koike et al. 1997; Mayer, Durward et al. 2002) and may be useful for screening purposes in large populations. It is commonly used to measure gastric emptying in critically ill adults (Heyland, Tougas et al. 1996; Heyland, Tougas et al. 1996) and even in pregnant women (Stanley, Magides et al. 1995). The safety and reliability of the test in neonatal population is not known. The newest technology in measuring gastric emptying is a technique involving a non-digestible capsule or SmartPill that can record luminal pH, temperature and pressure during gastrointestinal transit, thus allowing measurement of gastric emptying times (Kuo, McCallum et al. 2008). The data for gastric emptying times obtained with the SmartPill correlated well with data obtained with 57 Chapter 3 – Literature Review: Gastric Emptying scintigraphy. The technology does not involve any radioactivity and has been tested in healthy individuals. In summary, a number of different methods are available for the measurement of gastric emptying in humans, and all have some advantages and disadvantages. The method of choice will depend on whether solid or liquid meals are to be studied, level of precision required, degree of invasiveness that the subject will tolerate, ethical considerations, and the facilities available. 3.6 Methods and Factors Influencing Gastric Emptying in Neonates Until recently, few techniques to measure gastric emptying in neonates for clinical and research purposes were non-invasive, safe and reliable. Most data in preterm infants have been obtained using single aspiration technique (Husband and Husband 1969; Husband, Husband et al. 1970) or modified double sampling marker dilution technique (George 1968). Although the former method assessed the pattern of gastric emptying for an individual feed, the latter method however required repeated aspiration of gastric contents which may be unphysiological and not well tolerated in preterm infants (Ewer, Durbin et al. 1994) and thus obsolete due to availability of newer non-invasive techniques. Radionuclide methods using radioactive low-dose markers such as 99mTc sulfur colloid or with 99mTc phytate (Reddy, Deorari et al. 2000) mixed with milk is a reliable and reproducible technique to assess gastric emptying in infants. However the costs of a gamma camera and exposure to ionising radiation are major disadvantages in neonates and children. Various factors influence gastric emptying in neonates. Using marker dilution technique in newborns, Husband et al showed that 5% glucose and starch emptied faster than 10% glucose and glucose solution of comparable energy content (Husband and Husband 1969; Husband, Husband et al. 1970). Cavell using the same technique in healthy preterm infants showed that half the human milk and formula left the stomach in 25 and 51 minutes respectively (Cavell 1979). Other researchers also used marker dilution technique and demonstrated delayed gastric emptying to increased calorific density (Siegel, Lebenthal et al. 1984) and carbohydrate composition of feeds (Siegel, Krantz et al. 1985) and presence of respiratory distress syndrome (Victor 1975) and 58 Chapter 3 – Literature Review: Gastric Emptying congenital heart disease (Cavell 1981) in preterm infants. Feed osmolality (Hunt, Antonson et al. 1982; Siegel, Lebenthal et al. 1982) and the temperature of feeds (Costalos, Ross et al. 1979; Blumenthal, Lealman et al. 1980) did not appear to influence gastric emptying. Whereas the effects of fat content of feed (Sidebottom, Curran et al. 1983; Siegel, Krantz et al. 1985), phototherapy treatment (Blumenthal, Lealman et al. 1980; Costalos, Russell et al. 1984), and the posture of the infant (Victor 1975; Blumenthal and Pildes 1979) on gastric emptying was less clear. Using ultrasonography, Ewer et al confirmed Cavell’s finding that breast milk emptied faster than formula milk in preterm infants (Ewer, Durbin et al. 1994). The most likely reason for this observation was attributed to gut peptide content and composition of breast milk (Lucas, Sarson et al. 1980). ElHennawy et al failed to show change in gastric emptying rate, antroduodenal motor contractions and gastrointestinal transit times to intragastric erythromycin in feed intolerant preterm infants (ElHennawy, Sparks et al. 2003). The gastric emptying rate measured by 13C-octanoate breath test in preterm neonates was shown to be independent of milk amount (Pozler, Neumann et al. 2003). Similarly, gastric emptying was not affected by changes in feed osmolarity, volume, or energy density individually in preterm infants in a study by Ramirez et al. However reducing osmolality combined with increasing feed volume increased gastric emptying (Ramirez, Wong et al. 2006). Notably right lateral position increased gastric emptying in preterm infants (Omari, Rommel et al. 2004). Thus ultrasonography (Ewer, Durbin et al. 1994), (13)C-octanoic acid breath test (Pozler, Neumann et al. 2003) and epigastric impedance (Savino, Cresi et al. 2004) have gained popularity to monitor gastric emptying in premature infants and children both in research and clinical settings mainly due to the noninvasive nature of the technique and correlation with well-known techniques. The conflicting data on gastric emptying in new-born and preterm infants may be due to different methods of analysis and feed types used. 59 Chapter 4 – Literature Review: Feed Intolerance in Preterm Infants Chapter 4- Literature Review: Feed Intolerance in Preterm Infants 4.1 Scale of the Problem Provision of adequate nutrition for growth and development is a cornerstone of the care of preterm infants. However feed intolerance is a common problem and a major hurdle in optimising enteral nutrition in preterm neonates undergoing intensive care. Attempts to reach full enteral feeds in these infants are frequently limited by increased residual feeds in the stomach with or without other manifestations such as abdominal distension, bile stained aspirates, or lack of stooling. These manifestations in preterm infants are not exclusive to feed intolerance and can be experienced in other common clinical conditions such as infection and necrotising enterocolitis. Additionally the symptoms and signs of feed intolerance are difficult to quantify accurately and are prone to subjective variability. Thus the quoted incidence of feed intolerance in literature varies from up to 50% of ELBW infants (ElHennawy, Sparks et al. 2003) to 67% of VLBW infants (Gross 1983; Gross and Slagle 1993; Premji, Wilson et al. 1997). Under nutrition at critical stages of development may lead to short stature, organ growth failure, and later adverse behavioural and cognitive outcomes (Hay 2008). Therefore establishing full enteral feeds quickly in high risk preterm neonates has become a priority in neonatal intensive care (Patole, Rao et al. 2005). 4.2 Physiology of Gastric and Gastrointestinal Motility in Preterm Infants Mature gastrointestinal motility and migrating motor complexes depends on a complex interaction of neural and hormonal control mechanisms which are influenced by gestational age. By 34 weeks of gestation these MMCs are of variable length, with clear intervals and being increasingly propagated and except for the considerably shorter periodicity of 20-40 minutes, the MMC has adult characteristics at term gestation including interruption by feeding (Bryant, Buchan et al. 1982; Berseth 1989). 60 Chapter 4 – Literature Review: Feed Intolerance in Preterm Infants In new-born’s at birth cutaneous electrogastrography (EGG) (Abell and Malagelada 1988; Familoni, Bowes et al. 1991) demonstrated absence of normal slow waves and maturation process regulated by enteral feeding (Chen, Co et al. 1997; Liang, Co et al. 1998; Riezzo, Indrio et al. 2000). A recent study has shown mature EGG as early as 34 weeks gestation (Riezzo, Indrio et al. 2009). Fasting antral motor activity however is comparable in preterm and term neonates and the degree of antroduodenal coordination improves simultaneously towards term (Ittmann, Amarnath et al. 1992). 4.3 Definition of Feed Intolerance There are no universally agreed-upon criteria to define feed intolerance in preterm infants. Feed intolerance in preterm infants usually manifests as residual feeds in the stomach prior to the next scheduled feed and may be associated with vomiting, regurgitation, bile stained aspirates and abdominal distension (Akintorin, Kamat et al. 1997; Kliegman 1999; Rayyis, Ambalavanan et al. 1999; Dollberg, Kuint et al. 2000; Jadcherla and Kliegman 2002; Mihatsch, von Schoenaich et al. 2002). Of these, most clinicians consider vomiting, abdominal distension, and increased gastric residual volume as the ‘triad’ for defining feed intolerance. The literature definitions of feed intolerance in preterm infants mostly include an assessment of gastric residual volume (GRV) probably because it may indicate rapidity of gastric emptying. Furthermore majority of preterm and VLBW infants admitted to the neonatal unit need gastric tube placement and it is common practice to check the gastric residuals prior to the next scheduled feed. The significance of these GRV however may depend upon various factors (both systemic and local) including feeding method (continuous or bolus), feeding volume, feeding type (breast milk versus formula), feeding duration and birth weight and gestation of the infant. GRV are also dependent upon the technique of aspiration and body positions (Geraldo V 1997) and may be subject to variability. It is therefore not surprising that the definition of feed intolerance in preterm infants is varied in literature with several dynamic and fixed definitions based on assessment of prefeed residual volumes, the colour of these GRVs, and associated clinical manifestations such as abdominal distension, vomiting, blood in stool, and apnoea with bradycardia (Akintorin, Kamat et al. 1997; Rayyis, Ambalavanan et al. 1999; Dollberg, Kuint et al. 2000; 61 Chapter 4 – Literature Review: Feed Intolerance in Preterm Infants Mihatsch, von Schoenaich et al. 2002). Dynamic definitions of feed intolerance include GRV of > 20% (Dollberg, Kuint et al. 2000) of the previous 4 hour feed volume, > 50% (Akintorin, Kamat et al. 1997; Schanler, Shulman et al. 1999) or 20% (Rayyis, Ambalavanan et al. 1999; Dollberg, Kuint et al. 2000) of a 3 hour bolus feed volume or > 30% (Currao, Cox et al. 1988) or 20% (Wang, Hung et al. 1997; Dollberg, Kuint et al. 2000) of a 2 hour bolus feed volume, or as a total GRV > 10% of the previous day feed volume (Slagle and Gross 1988). Fixed definitions of feed intolerance include GRV > 2ml of undigested formula (Silvestre, Morbach et al. 1996), > 2ml or any bilious coloured aspirate (Dunn, Hulman et al. 1988), or > 3 ml/kg or any green coloured aspirate (Meetze, Valentine et al. 1992). Critical assessment of these publications revealed small sample size and variable birth weights and gestations, arbitrarily developed definitions based on consensus among the study staff and the attending neonatologists (Akintorin, Kamat et al. 1997), use of variable dilutions of formula feeds and hypocaloric feeds (Currao, Cox et al. 1988; Silvestre, Morbach et al. 1996), variable feed types, duration, and method of feeding (all studies), and prolonged period of nil by mouth and TPN use with variable feed advancement rate (Slagle and Gross 1988). All the studies used excessive GRV, either determined by % of the previous feeding volume or an absolute volume, as a proxy for feeding intolerance and the authors provided no physiological basis or demonstrated correlation with the known measures of gastric emptying or gastrointestinal motility or relevance to clinical outcomes. Even when definition seemed comprehensive the clinical significance of each of the criteria for feeding intolerance was not determined (Mihatsch, von Schoenaich et al 2002; Dollberg, Kuint et al 2000; Rayyis, Ambalavanan et al 1999; Akintorin, Kamat et al 1997). A study which did attempt to investigate the impact of high GRV and colour in a cohort of ELBW infants showed that threshold GRV of 2ml/3ml and green, milky, or clear GRV had no significant impact on feeding volume on day 14 (Mihatsch, von Schoenaich et al. 2002). However, the median feeding volume of 103 (0-166) ml/kg birth weight on day 14 in the study was far below volumes aimed for in current practice which may have contributed to gut hypomotility. Additionally, the results cannot be generalised for infants fed at an incremental rate > 12ml/kg/day or with a different formula. 62 Chapter 4 – Literature Review: Feed Intolerance in Preterm Infants On the other hand, threshold or % prefeed GRV was assessed against objectives measures of gastric emptying in critically ill children and adults (Mayer, Durward et al. 2002; ElHennawy, Sparks et al. 2003; Gomes, Hornoy et al. 2003; Pozler, Neumann et al. 2003). Using GRV > 125% of previous 4 hour feed volume or > 150 ml as indicative of feed intolerance, Mayer et al showed correlation with delayed gastric emptying measured by paracetamol absorption test. However the study sample sizes were small (Mayer, Durward et al. 2002; ElHennawy, Sparks et al. 2003; Gomes, Hornoy et al. 2003; Pozler, Neumann et al. 2003) and the study cohort was of wide ranging age and weight (Mayer, Durward et al. 2002). Thus the definition of feed intolerance in the literature varies and is arbitrarily based mainly on an assessment of prefeed gastric residual volumes, the colour of these GRVs, and the associated clinical manifestation. Although a high preprandial GRV or green GR are generally regarded as significant markers of feed intolerance, its clinical significance is yet to be determined. It is unclear if they are predictive of serious disease such as necrotising enterocolitis, a delayed time to achieve full enteric alimentation, or developmental physiologic expectations when feeding the preterm infants. 4.4 Patho-Physiology of Feed Intolerance Feed intolerance is rarely observed in well term infants most likely due to controlled gastrointestinal motility which matures as gestational age increases (Tomomasa, Itoh et al. 1985; Berseth 1989; Berseth 1990; Berseth 1996). The gastrointestinal tract has formed and has completed its rotation back into the abdominal cavity by 10 weeks of gestation. The fetus can swallow amniotic fluid by 16 weeks and gastrointestinal motor activity is present before 24 weeks but organized peristalsis is not established until 29-30 weeks. At 32-34 weeks coordinated sucking and swallowing develops and by term the fetus swallows about 150 ml/kg/day of amniotic fluid containing carbohydrates, protein, fat, electrolytes, immunoglobulin’s and growth factors which plays an important role in development of gastrointestinal function. Preterm birth interrupts this development. In the postnatal period the lack of enteric intake leads to decreased circulating gut peptides, slower enterocyte turnover and nutrient transport, decreased bile acid secretion; even when nutrients are provided 63 Chapter 4 – Literature Review: Feed Intolerance in Preterm Infants parenterally. There is increased susceptibility to infection due to impaired barrier function by intestinal epithelium, lack of colonization by normal commensal flora and colonization by pathogenic organisms. Physiologic studies have shown a lack of the propagative phase III of the MMC in the duodenum of preterm infants less than 32 weeks gestational age (al Tawil and Berseth 1996). Additionally, very immature infants also have poor gastroduodenal coordination and excessive quiescence in motor activity (Jadcherla and Kliegman 2002) and nonpropagated contractions of the gastrointestinal tract. Furthermore, control systems of gastrointestinal motility are immature in preterm animals because interstitial cells of Cajal (ICC), nerve, and muscle control systems are not fully differentiated (Daniel and Wang 1999; Jadcherla and Kliegman 2002). Therefore immaturity of the gastrointestinal tract due to premature birth has been proposed as the main cause of feed intolerance in preterm infants (Ng and Shah 2008). Prefeed gastric residual volumes (GRV) and bilious residuals may reflect poor gastric emptying, duodeno-gastric reflux, and gastroduodenal hypomotility (Jadcherla and Kliegman 2002). Although not proven, it is likely that colonic motility is also slow and delayed in VLBW infants as supported by delayed stooling pattern, and emesis in bowel obstruction or functional ileus (Jadcherla and Kliegman 2002). Although historically hypoglycaemia has been the clinical concern, hyperglycaemia is also a well-documented problem in the perinatal period in VLBW infants. This hyperglycaemia is a marker of insulin resistance and relative insulin deficiency and may reflect the prolonged catabolism observed in VLBW infants (Beardsall and Dunger 2007). Intrauterine growth retardation is also associated with increased fasting insulin levels and insulin resistance (Mericq 2006; Mericq 2006). These infants are unable to suppress glucose production within a large range of glucose and insulin concentrations, insulin secretory response is inappropriate, insulin processing is immature and there is an increased ratio of the glucose transporters Glut-1/Glut-2 in fetal tissues, which limits sensitivity and hepatocyte reaction to increments in glucose/insulin concentration during hyperglycaemia (Mericq 2006). Although unproven, it is plausible that relative insulin resistance in preterm VLBW may induce 64 Chapter 4 – Literature Review: Feed Intolerance in Preterm Infants hyperamylinemia which may be responsible for feed intolerance by delaying gastric emptying. In conclusion, prematurity may predispose to feed intolerance due to immaturity of gastrointestinal motor activity (Bisset, Watt et al. 1988; Berseth 1989; Reddy, Deorari et al. 2000; Patole, Rao et al. 2005), and delayed gastric emptying and whole intestinal transit (Gupta and Brans 1978; Cavell 1982; Ernst, Rickard et al. 1989). Relative insulin resistance and hyperamylinaemia may also be responsible for feed intolerance in these neonates which is an emerging theory and hypothesis and subject of investigation of this dissertation. 4.5 Management of Feed Intolerance A range of management strategies currently exists for preventing and/or minimizing feed intolerance in preterm neonates reflecting the dilemma surrounding the definition and significance of signs of feed intolerance due to gastrointestinal hypomotility on one end and the fear of NEC on the other end of the spectrum. Additionally, the relationship of feed intolerance and gastroesophageal reflux is unclear and therefore often used interchangeably. Furthermore due to the absence of robust diagnostic tests authors have resorted to surrogate markers of gastrointestinal motility such as gastric residuals and time to reach full enteral feeds to assess efficacy of therapeutic interventions. The following paragraphs provide critical appraisal of current literature on the management of feed intolerance in preterm infants. Prokinetic agents are often used in an attempt to overcome the functional immaturity by speeding up gastric emptying and increasing small intestinal motility. Until July 2000, cisapride was the mainstay of therapy in preterm infants with feed intolerance and/or gastro-oesophageal reflux. Due to reports of prolongation of the QT interval with occasional fatal cardiac arrythmias and sudden death, cisapride was taken off the market and restricted to a limited access programme supervised by a paediatric gastroenterologist (Gounaris, Costalos et al. 2010; Maclennan, Augood et al. 2010). Metoclopromide and domperidone, dopamine antagonists, started to be widely prescribed soon after (Clark, Bloom et al. 2006; Hibbs and Lorch 2006) although its efficacy was in doubt. A systematic review of metoclopramide use did not show benefit but 65 Chapter 4 – Literature Review: Feed Intolerance in Preterm Infants reported the possibility of adverse effects such as irritability, dystonic reactions, oculogyric crisis, drowsiness, apnea and emesis (Putnam, Orenstein et al. 1992; Hibbs and Lorch 2006). There have been no adequately powered controlled trials of metoclopramide use in preterm neonate. Similarly efficacy data of domperidone in preterm infants is lacking. The few studies with domperidone are old and methodologically poor. In contrast the side effects profile is similar to those of cisapride with prolonged QT interval, sudden death and theoretical risk of neurological adverse effects. A recent small crossover study of domperidone use in 22 preterm infants showed faster gastric emptying rate measured by ultrasound antral cross sectional area half-value time (in minutes) n the domperidone group (Gounaris, Costalos et al. 2010). However the study did not report on important clinical correlates of efficacy and long term safety. There have been no controlled trials of domperidone use in preterm infants. A plethora of publications soon followed on erythromycin use in the prevention and treatment of feeding intolerance in preterm infants. Erythromycin is a motilin agonist that exerts its prokinetic effect by stimulating propagative contractile activity in the interdigestive phase. Although previous studies demonstrated variable efficacy, a meta-analysis (three prevention and seven treatment studies) concluded that there is insufficient evidence to recommend the use of erythromycin in low or high doses for preterm infants with or at risk of feeding intolerance (Ng and Shah 2008). Furthermore the same author concluded that although high dose regimens as a rescue therapy for established gastrointestinal dysmotility have consistently shown clinical benefit, further research is warranted to justify its widespread use due to the theoretical risks of prolonged antibiotic use (Ng et al 2011) and hypertrophic pyloric stenosis. Clinical trials followed to investigate interventions to promote intestinal maturation and enhance feeding tolerance and decrease time to reach full enteral feeding independent of parenteral nutrition. Systematic review of nine trials did not provide any evidence that early trophic feeding affected feed tolerance or growth rates in VLBW infants (Bombell and McGuire 2009). Trials of supplementation of breast milk or formula milk with enzyme or hormone to improve short and long term effects of feed intolerance were also conducted. A randomized trial did not provide evidence of significant benefit to preterm infants 66 Chapter 4 – Literature Review: Feed Intolerance in Preterm Infants from adding lactase to their feeds (Tan-Dy and Ohlsson 2005). A study of eight preterm infants who were given 4 U/kg/day insulin enterally from 4 to 28 days of age showed higher lactase activity and less feed intolerance (30% shorter time to full enteral feeds; fewer gastric residuals per infant) than the matched historical controls (Shulman 2002). However further large controlled studies are warranted before its use in preterm population. Recent literature abounds in the study of probiotics, prebiotics, or synbiotics in preterm infants primarily looking to improve growth and reduce mortality due to NEC and infections. Some of these trials reported on their effect on feed tolerance and/or days to reach full enteral feeds as a surrogate marker of feed tolerance. L. sporogenes supplementation at the dose of 350,000,000 c.f.u/day was not effective in reducing the incidence of death or NEC in VLBW infants; however, it could improve the feeding tolerance (Sari, Dizdar et al. 2011). Although the time to reach full enteral feeds was significantly shorter in the early enteral probiotic group than in controls (Deshpande, Rao et al. 2007) an updated meta-analysis did not show this effect which was attributed to the significant heterogeneity among the included trials (Deshpande, Rao et al. 2010). The results of ProPrems trial (Garland, Tobin et al. 2011)- a multi-centre, randomised, double blinded, placebo controlled trial investigating supplementing preterm infants born at < 32 weeks' gestation weighing < 1500 g, with a probiotic combination (Bifidobacterium infantis, Streptococcus thermophilus and Bifidobacterium lactis) is awaited which in addition is investigating time to reach full enteral feeds. Supplementation with Bifidobacterium longum and Lactobacillus rhamnosus may improve the gastrointestinal tolerance to enteral feeding in infants weighing >1000 g (Rouge, Piloquet et al. 2009). Enteral supplementation of a prebiotic mixture consisting of neutral and acidic oligosaccharides decreased stool viscosity and stool pH with a trend towards increased stool frequency without demonstrating benefit on feed tolerance (Westerbeek, Hensgens et al. 2011). Evidence is still emerging with regards to the strain type, supplementation to breast or formula and dose, frequency and duration of prebiotics and probiotics in relation to its effects on feed intolerance and other important clinical outcomes including safety data. The ESPGHAN Committee on nutrition considers that the supplementation of formula with probiotics and/or prebiotics does not raise safety concerns but 67 Chapter 4 – Literature Review: Feed Intolerance in Preterm Infants there is insufficient data to recommend routine supplementation and therefore is an important field of research (Braegger, Chmielewska et al. 2011). In clinical practice, failure of pharmacological intervention may necessitate trial of transpyloric feeding as a last resort. However transpyloric route cannot be recommended for preterm infants (McGuire and McEwan 2007) due to adverse effects such as greater incidence of gastrointestinal disturbances and some evidence of increased mortality. The fallback position when all intervention fails is to continue total parenteral nutrition until the feed intolerance settles. In conclusion management of feed intolerance in preterm infants is varied reflecting the dilemma surrounding the definition of feed intolerance. They include pharmacological and biological interventions the efficacy of which is variable. Further trials are awaited for evidence based management of this common condition. 68 Chapter 5 – Literature Review: Gestational Diabetes Mellitus Chapter 5- Literature Review: Gestational Diabetes Mellitus 5.1 Definition of Gestational Diabetes Mellitus Gestational diabetes mellitus (GDM) is defined as any degree of glucose intolerance with onset or first recognition during pregnancy (Metzger and Coustan 1998) 5.2 Scale of the Problem 3-10% of all pregnancies (Hoffman, Nolan et al. 1998) experience GDM. This number may rise significantly in the next decade as the current significantly overweight paediatric population heads into their child-bearing years. GDM is not only associated with substantial rates of maternal adverse effects but also periconceptional, fetal, neonatal, and long term morbidities (Nold and Georgieff 2004). Long-term adverse health outcomes reported among infants born to mothers with gestational diabetes (IMDM) include sustained impairment of glucose tolerance (Silverman, Metzger et al. 1995), subsequent obesity (Petitt, Bennett et al. 1985), and impaired intellectual achievement (Rizzo, Metzger et al. 1997; Rizzo, Silverman et al. 1997). 5.3 Physiology of Insulin Secretion Glucose and several amino acids stimulate insulin secretion under physiologic conditions. The rate of insulin secretion is dependent on the ratio of ATP to ADP within the pancreatic beta cell. An increase in the intracellular ATP/ADP ratio activates membrane ATP-sensitive potassium-dependent channels (KATPs) leading to closure of the potassium channel and depolarization of the cell membrane allowing influx of calcium through voltage-gated calcium channel resulting in insulin secretion. The rate of glucose entry into the beta cell exceeds its oxidation rate which is facilitated by a glucose transporter. The first rate-limiting step in glycolysis which is conversion of glucose to glucose-6phosphate by the enzyme glucokinase regulates the rate of glucose oxidation and subsequent insulin secretion. 69 Chapter 5 – Literature Review: Gestational Diabetes Mellitus 5.4 Physiology of Gestational Diabetes Mellitus Pregnancy is a physiologic condition in which maternal glucose is the main energy substrate for intrauterine growth (Combs, Gunderson et al. 1992). Early in pregnancy, increased levels of both oestrogen and progesterone act as counter-regulatory hormones affecting glucose homeostasis. As pregnancy progresses, human placental lactogen released by the syncytiotrophoblast leads to lipolysis and the subsequent release of glycerol and fatty acids which reduces maternal use of glucose and amino acid, thus preserving these substrates for the fetus. As placental growth and pregnancy continue, maternal insulin needs increase by 30% due to the release of increasing amounts of counter-insulin factors. Glucose and amino acids readily traverse the placental membrane while insulin is unable to cross materno-fetal barrier in physiological concentrations. In pregnancies complicated by diabetes, high concentrations of glucose, particularly after a meal, give rise to an increased nutrient transfer to the fetus. This results in beta-cell hyperplasia, stimulating an increased release of insulin which adversely influences birth weight with its pregnancy complications (Jovanovic-Peterson, Peterson et al. 1991; Combs, Gunderson et al. 1992). 5.5 Strategies to Hyperinsulinaemia Prevent Fetal and Neonatal The Diabetes Prevention Program randomised clinical trial showed reduction of diabetes incidence in high-risk adults by 58% with intensive lifestyle intervention and by 31% with metformin (Knowler, Barrett-Connor et al. 2002) the benefits of which persisted for at least 10 years (Knowler, Fowler et al. 2009). A randomised trial of dietary advice, blood glucose monitoring, and insulin therapy or routine care in women between 24 and 34 weeks' gestation who had gestational diabetes showed significantly lower serious perinatal complications such as death, shoulder dystocia, bone fracture, and nerve palsy (Crowther, Hiller et al. 2005). Continuous glucose monitoring during pregnancy is associated with improved glycaemic control in the third trimester, lower birth weight, and reduced risk of macrosomia (Murphy, Rayman et al. 2008). Compared to normal pregnant women in the low-glycaemic index group, women in the moderate-to-high glycaemic index group gave birth to infants who were 70 Chapter 5 – Literature Review: Gestational Diabetes Mellitus heavier and had a higher birth centile, a higher ponderal index, and a higher prevalence of large-for-gestational age (Moses, Luebcke et al. 2006). Low– glycaemic index diet for women with gestational diabetes also halved the number needing to use insulin, with no compromise of obstetric or fetal outcomes (Moses, Barker et al. 2009). Both intensive lifestyle and metformin were highly effective in delaying or preventing diabetes in women with impaired glucose tolerance test and a history of gestational diabetes (Ratner, Christophi et al. 2008). On the other hand, a multicentre randomised control trial of treatment of mild gestational diabetes did not significantly reduce the frequency of a composite outcome that included stillbirth or perinatal death and several neonatal complications; however it did reduce the risks of fetal overgrowth, shoulder dystocia, caesarean delivery, and hypertensive disorders (Landon, Spong et al. 2009). No substantial maternal or neonatal outcome differences were found with the use of glyburide or metformin compared with use of insulin in women with GDM (Nicholson, Bolen et al. 2009). Fat mass, body mass index, ponderal index, skinfold sum, arm fat area, and cord concentrations of biomarkers were not different in infants of women treated for gestational diabetes with glyburide or insulin (Lain, Garabedian et al. 2009). In women with gestational diabetes at 20-33 weeks of gestation, metformin (alone or with supplemental insulin) as compared with insulin was not associated with increased perinatal complications (Rowan, Hague et al. 2008). Glucose control in women with gestational diabetes mellitus treated with metformin and/or insulin was strongly related to outcomes. The lowest risk of complications was seen when fasting capillary glucose was <4.9 mmol/l (mean ± SD 4.6 ± 0.3 mmol/l) compared with 4.9–5.3 mmol/l or higher and when 2-h postprandial glucose was 5.9–6.4 mmol/l (6.2 ± 0.2 mmol/l) or lower (Rowan, Gao et al. 2010). Although treatment of GDM substantially reduced macrosomia at birth, it did not result in a change in BMI at age 4-5 years (Gillman, Oakey et al. 2010). Thus strict control of blood sugar during pregnancy in GDM prevents fetal and neonatal hyperinsulinaemia and related complications but may not help in reversing intrauterine programming resulting in future adverse outcomes. 71 Chapter 6 – Literature Review: IMDM Chapter 6- Literature Review: IMDM 6.1 Scale of the Problem Currently, 3-10% of pregnancies are affected by abnormal glucose regulation and control. Of these cases, 80-88% are related to gestational diabetes mellitus. Of mothers with preexisting diabetes, 35% have been found to have type 1 diabetes mellitus, and 65% have been found to have type 2 diabetes mellitus. Notably infants born to mothers whose pregnancy was complicated by diabetes (gestational and types 1 and 2 diabetes) have experienced nearly 30-fold decrease in morbidity and mortality rates since the development of specialized maternal, fetal, and neonatal care for women with diabetes and their offspring. However adverse neonatal outcomes are still 3-9 times greater in infants born to mothers with preexisting types 1 and 2 diabetes compared to those of nondiabetic mothers (Yang, Cummings et al. 2006). In mothers with insulindependent diabetes, the stillbirth and perinatal mortality rate is 5 times the rate in the general population, and neonatal and infant mortality rates are 15 times and 3 times the rate in the general population, respectively. Major congenital malformations are found in 5-9% of affected infants and account for 30-50% of perinatal deaths of infants of mothers with gestational diabetes. IMDM (gestational, types 1 and 2 diabetes) are 3 times more likely to be born by caesarean delivery, twice as likely to suffer serious birth injury, and 4 times as likely to be admitted to a neonatal intensive care unit. In addition they are at increased risk of morbidity and mortality related to the abnormalities in fetal growth (either overgrowth or undergrowth), birth injuries, hypoglycaemia, hypocalcaemia, feed intolerance, polycythaemia and thrombocytopenia, jaundice, prematurity, respiratory distress syndrome, asymmetric septal hypertrophy, congenital malformations and intrapartum asphyxia. 6.2 Physiology of Fetal Hyperinsulaemia The physiology of fetal hyperinsulinaemia in mothers with gestational diabetes is explained by Pedersen hypothesis which states that maternal hyperglycemia results in fetal hyperglycemia as glucose readily traverses the placenta 72 Chapter 6 – Literature Review: IMDM (Pedersen 1977). Before 20 weeks' gestation, fetal islet cells are not capable of responsive insulin secretion. After 20 weeks’ gestation, the fetus has a functioning pancreas and is responsible for its own glucose homeostasis. Therefore fetal hyperglycemia induces fetal pancreatic beta-cell hyperplasia with resultant hyperinsulinaemia. In addition, proinsulin hormones such as insulin like growth factor-1 [IGF-1] and insulin like growth factor–binding protein3 [IGFBP-3] act as growth factors that, in the presence of increased fetal amino acids result in fetal macrosomia noted as early as 24 weeks' gestation. The combination of hyperglycaemia and insulin increases fetal fat and glycogen stores, resulting in weight increases marked by hepatosplenomegaly and cardiomegaly. Chronic fetal hyperglycaemia and hyperinsulinaemia increase the fetal basal metabolic rate and oxygen consumption, leading to a relative hypoxic state. The fetus responds by increasing oxygen-carrying capacity through increased erythropoietin production, potentially leading to polycythaemia. Prior to birth, elevated insulin levels may inhibit the maturational effect of cortisol on the lung, including the production of surfactant from type 2 pneumocytes. This puts the fetus at risk for developing respiratory distress syndrome after birth at a gestational age when normal lung function is expected. Withdrawal of the transplacental supply of glucose after birth leads to a precipitous drop in the concentration of glucose; many require infusion of large quantities of glucose to maintain normal blood glucose levels. 6.3 Feed Intolerance in IMDM Feed intolerance occurs in up to 37% of IMDM (Kitzmiller, Cloherty et al. 1978) and a key reason for prolongation of hospital stay and parent-infant separation (Lee-Parritz 2004). It is often present in the absence of prematurity, respiratory distress, and polyhydramnios, and the incidence is similar among large-forgestational-age and appropriate-for-gestational-age infants (Lee-Parritz 2004). This frequently observed problem of feed intolerance in IMDM is overshadowed by previously described morbidities (section 5.1) and therefore not widely reported. The management of feed intolerance in IMDM relies on tube feeding with or without intravenous dextrose solution until the infant establishes to demand oral feeds. The conservative approach to management may be due to 73 Chapter 6 – Literature Review: IMDM the fact that the aetiology and pathophysiology of feed intolerance in IMDM is yet to be determined. 6.4 Proposed Patho-Physiology of Feed intolerance in IMDM Elevation of plasma amylin levels has been described in pregnant women with gestational diabetes mellitus (Ludvik, Kautzky-Willer et al. 1997). Insulin resistance and hyperinsulinaemia probably due to hyperleptinemia has been demonstrated in IMDMs (Vela-Huerta, San Vicente-Santoscoy et al. 2008). It is plausible that this hyperinsulinaemia and insulin resistance may also predispose to hyperamylinaemia which may have a role to play in the pathogenesis of feed intolerance in IMDMs. This plausible mechanism and hypothesis is yet to be proven and a subject of investigation of this dissertation. 74 Chapter 7 – Overview of Project Plan Chapter 7- Overview of Project Plan 7.1 General Design The overall project plan (Figure 16) was to test the hypothesis that amylin would be raised in neonates with clinical signs of feed intolerance. Figure 16. Project plan Gantt chart 75 Chapter 7 – Overview of Project Plan As there was no published information regarding amylin and its concentrations in the newborn, the first part of the study was to establish normative values of amylin at birth and postnatal period in term and well preterm infants. Measurement of amylin levels at more than one time point during the first weeks of life following birth would help in the assessment of its trend and decay with time. It was also essential to identify two or more time points after birth that would allow for blood collection to be undertaken without causing discomfort and pain to the study participants. Umbilical cord blood at birth and Guthrie blood on postnatal day 5 were identified as practically feasible time points requiring minimal or no intervention in the mother and the baby. This approach required collection of blood both in the hospital (from umbilical cords and at the time of Guthrie of in-house babies) and community setting (at the time of Guthrie of those babies discharged home from the hospital before day 5). A pilot study of community blood collection identified unacceptable compromises to the sampling process and maintenance of cold chain. Therefore the study plan was amended to abandon community blood collections. Umbilical cord blood could be collected from discarded cords immediately after birth without causing pain or discomfort to the mother and the baby. The blood for Guthrie test is collected from a heel-prick sample and is offered to all infants in the UK with the aim of screening for up to five disorders typically on day 5 after birth. Thus it was feasible to collect blood for amylin during routine Guthrie test minimizing pain and discomfort. A third time point in the 2nd week of postnatal life although necessary was deemed impractical due to logistical reasons around blood sampling and ethical concerns of blood collection purely for study purposes. Originally, the second part of the project was to determine amylin levels in feed intolerant infants of mothers whose pregnancy was complicated by diabetes mellitus. To address the hypothesis that amylin levels are raised in IMDM with feed intolerance, a case-control study of feed intolerant and feed tolerant IMDM was considered. However an audit showed that not all IMDM were admitted to the neonatal intensive care or special care baby unit. Additionally, not all admitted IMDM required gastric tube placement as part of their routine care. Insertion of gastric tube to measure prefeed gastric residual volumes was essential to define feed intolerance. This meant designing a new guideline for 76 Chapter 7 – Overview of Project Plan the purpose of the study with inherent ethical issues. Therefore case-control study design was abandoned in favor of a cohort or prospective observational study of IMDM as an appropriate alternative methodology to address the hypothesis. Thus if amylin levels were observed to be raised in IMDM then it was not necessary to provide proof of concept as amylin was shown in previous studies to be a potent inhibitor of gastric emptying. Measurement of amylin levels at more than one time point during the first weeks following birth would help analyse its trend and decay with time. It was therefore necessary to identify at least two points in time following birth of the IMDM for the bloods to be collected causing minimal discomfort and pain. Umbilical cord blood at birth and Guthrie blood on postnatal day 5 were identified as practically feasible time points requiring minimal or no intervention in the mother and the baby. A third time point for opportunistic blood collection was also agreed when IMDM were admitted to the neonatal intensive care unit or transitional care unit for IMDM related problems and needing routine blood sampling. The third part of the study was to test the hypothesis that amylin was raised in preterm infants with feed intolerance. A cohort study of preterm infants with feed intolerance alone may not conclusively address the hypothesis as measurement of gastric emptying times in addition to gastric residual volumes would have to be incorporated into the study design. Review of currently available tests to measure gastric emptying times at both the study centres were not considered to be practical, feasible and for some tests to be ethical: The expertise to measure gastric emptying time using currently available tests such as scintigraphy, ultrasonography, bio impedence, and 13C-octanoate breath test was not available at Doncaster General Hospital. Similarly bioimpedence technology and 13C-octanoate breath test were not available at Jessop Wing. Additionally the research team felt that serial ultrasonographic assessment of ACSA at Jessop Wing was not practical and feasible due to non-availability of radiology expertise out of hours and on weekends. Although gamma camera was available within the hospital building, the need to transfer the infants to the radiology department for this to be undertaken would put strain on existing resources in addition to a small risk of exposure to radiation. The question regarding safety of paracetomol in preterm infants and need for serial blood sampling for paracetomol absorption test would raise ethical concerns. A case77 Chapter 7 – Overview of Project Plan control study was therefore adopted to answer the hypothesis. Preterm infants less than 34 weeks are routinely admitted to the neonatal unit and most of these infants require naso/orogastric tube placement and routine blood sampling as part of their care. 7.2 General Study Configuration and Methodology The study project involved observation of blood concentration of amylin in neonatal population and therefore did not require randomisation or blinding. The project was conducted simultaneously as three interrelated studies (Figure 17 & Figure 18) for operational reasons and initially required a single-centre research ethics application. 78 Chapter 7 – Overview of Project Plan Figure 17. Study plan Gantt chart 79 Chapter 7 – Overview of Project Plan PROJECT TITLE: AMYLIN PEPTIDE: AN ASSOCIATION WITH FEED INTOLERANCE IN PRETERM NEONATES AND IMDM Study A Amylin levels at birth and postnatal day 5 in healthy preterm and term infants Study B Amylin levels at birth and postnatal day 5 in preterm and term IMDM Study C Amylin levels in preterm infants with feed intolerance Single Centre Cohort Study Two Centre Case-Control Study Labour ward, postnatal wards, Neonatal intensive care unit Jessop wing, Royal Hallamshire Hospital Inclusion ≥ 24 week gestation infants born without major antenatal or labour complications Neonatal intensive care unit Jessop wing, Royal Hallamshire Hospital & Doncaster General Hospital Inclusion ≥ 24 weeks gestation born to mothers with diabetes mellitus (gestational, insulin or non-insulin dependent) Inclusion 24-36 weeks gestation admitted to intensive care unit and requiring gastric tube feeding Prefeed GRV > 50% on 2 consecutive occasions Yes=nTOL No=TOL Matched for birthweight, gestation, postnatal age Parental consent 0.5-1.0 ml blood collected from discarded umbilical cord, during routine collection and Guthrie 0.5-1.0 ml blood collected within an hour Plasma stored at -70 degrees ELISA Amylin levels Insulin levels (where possible) Figure 18. General study methodology 80 0.5-1.0 ml blood collected at the next routine collection Chapter 7 – Overview of Project Plan These three studies were planned to complete in 12 months; 10 months for recruitment and sample collection; and 2 months for completion of laboratory and data analysis. Study A was a single centre prospective observational study in well preterm and term infants to determine normal ranges of amylin in umbilical cord (birth) and Guthrie (postnatal day 5) blood. Study B was a single centre prospective observational study of IMDM to determine amylin levels in umbilical cord (birth), during routine blood sampling (neonatal intensive care stay) of admitted babies and Guthrie (postnatal day 5). Study C was a two centre case-control study to determine amylin levels in preterm infants admitted to the neonatal intensive care unit who subsequently developed feed intolerance and compared to amylin levels in birth weight, gestation and postnatal day of life matched preterm infants without feed intolerance. Some of these preterm infants were also recruited to study A. A second centre was enrolled to ensure the study was conducted to time lines. There were no modifications to the treatment of care received by the study infants. Blood samples for the study were obtained only when blood was being collected for routine clinical monitoring as this approach minimised discomfort to the infant. Doctors, nurses and midwifes working in the neonatal unit and labour ward performed routine blood sampling. Further training was imparted by the research team to nurses, midwives and doctors in blood collection, processing and storage. The transportation of the stored plasma to the laboratory was performed under controlled conditions using liquid nitrogen to maintain the cold chain. 7.3 Ethics Consideration The study was conducted according to the standards of International Conference on Harmonisation Good Clinical Practice Guideline, Research Ethics Committee regulations, Royal Hallamshire Hospital’s policies and procedures, and any applicable government regulations. The study protocol and any amendments were submitted to a properly constituted Ethics Committee (REC) for approval of the study conduct. The 81 Chapter 7 – Overview of Project Plan REC requested minor amendments to the protocol (Appendix 4 & Appendix 5), information leaflets (Appendix 6, Appendix 7 & Appendix 8), and adverts (Appendix 11, Appendix 12 & Appendix 13) which were incorporated before final approval in writing. All parents/guardians were provided with an information sheet (Appendix 6, Appendix 7, Appendix 8) describing the elements of this study, with sufficient information for them to make an informed decision about their infant participating in this study. The parents/guardians completed and signed a consent form to indicate that they were giving consent for their infant to participate (Appendix 9 & Appendix 10). It was made clear to the parents/guardians that the study will offer no direct benefit to their own child. Additionally they were free to withdraw their infant from the study at any time. Participants were discontinued from study at any time for safety reasons as judged by the investigator or clinician responsible for the child and when parents withdrew consent. The possible ethical considerations arising from the conduct of the study were: (1) Blood collection from umbilical cord attached to the baby. An amendment was accepted to collect the blood samples from the discarded umbilical cord to prevent discomfort, bleeding and possible parent-baby separation. (2) Consenting pregnant mothers in active labour. The ethics committee agreed for the blood to be collected from umbilical cords immediately after birth of the baby without prior consent. The study investigators may approach the mothers after delivery for consent and collected blood sample discarded if consent is refused. To circumvent this problem and to fulfil research governance obligations, the research team decided to amend the study plan to recruit mothers that were admitted for elective induction of labour or caesarean section and umbilical cord blood collected after informed and written consent. (3) The collection and possession of human tissue. The umbilical cords were disposed immediately after blood collection according to hospital policy. The left over plasma was disposed immediately after analysis. 82 Chapter 7 – Overview of Project Plan (4) Blood collection from preterm infants and volume of blood. Blood samples were obtained during times when blood was collected for routine clinical monitoring. This approach minimised any extra discomfort to the infant. All blood handling was performed as per guidance for the minimization of infection risk to both participants and investigators. Not more than 2 ml of blood was collected from any infant during the course of the study which was within the safe limit. 7.4 Data Handling and Record Keeping Confidentiality: Information about study subjects was kept confidential and managed according to the requirements of the Data Protection Act, NHS Caldicott Guardian, The Research Governance Framework for Health and Social Care and Ethics Committee Approval. The chief investigator acted as custodian of the data. Each participant was assigned a unique study number, allocated for use on data collection forms, other documentation and the electronic database. The data collection form was the only form to contain identifying data and were treated as confidential documents and held securely in accordance with the relevant regulations. Access was restricted to those personnel approved by the chief investigator. Blood samples were sent to the laboratory for analysis labelled only with the unique identification number. Data were anonymised on a computer database for the study by using the subject identification number only and this anonymised data was used for the statistical analyses specific to this study. Data Collection Forms or Case Report Forms (Appendix 14, Appendix 15 & Appendix 16) recorded participants’ biochemistry and demographics. Relevant information was retrieved from an electronic patient database and patient notes. Infant data included demographic variables such as birth weight, gestation, and gender, and postnatal age, date of blood collection which were recorded for all neonates. Maternal data included medical, antenatal, and labour history. For Study C the neonatal data also included ventilation days, % GRV just prior to amylin sampling, time from initiation to full enteral feeds (defined as 150 ml/kg per day) and time to discharge. Additionally, inflammatory markers such as C83 Chapter 7 – Overview of Project Plan reactive protein, white cell count, and neutrophil cell count were also recorded nearest to the date of blood sampling. Incomplete datasets from subjects discontinued from the study were not used. Records Retention: Documents were stored in a locked cabinet with limited access. Good Clinical Practice Guidelines stipulated that for patients involved in clinical trials documentation should be retained for 25 years after conclusion of treatment. 7.5 Benefits and Risks There would be no health benefit to the participants themselves. The results obtained from this study will help to establish normative amylin data in healthy preterm and term infants. The study may also help unravel the pathophysiology of feed intolerance which may lead to intervention studies in IMDM and preterm infants. Blood was collected from discarded umbilical cords for study A and B. No more than a total of 2.0 ml of blood was collected during the whole study from each baby in study A, B & C. This is also an acceptable amount to take from a pre-term baby (Directive 2008). 7.6 Funding The investigators received £5000 from Jessop Wing Baby Fund to complete the three studies. Three thousand and five hundred pounds was returned as only £1500 was spent on the entire project. The money was utilized for the following: Amylin Kits £1440 Serum tubes £50 Technical staff time £0 Trasylol £10 84 Chapter 8 – Clinical Study A Chapter 8Clinical Study A: Umbilical & Guthrie Amylin in Preterm and Term Infants 8.1 Introduction The aim of this study was to determine the normal range of serum amylin levels in healthy preterm and term neonates at birth and postnatal day 5. This approach required collection of blood both in the hospital (from umbilical cords and at the time of Guthrie of in-house babies) and community setting (at the time of Guthrie of those babies discharged home from the hospital before day 5). A pilot study of community blood collection however identified unacceptable compromises to the sampling process and maintenance of cold chain. Therefore the study aim was amended in favor of blood collection of those infants who remain in house on postnatal day 5. 8.2 Methodology 8.2.1 Study Design & Setting This single centre prospective observation study was conducted at the University Hospitals of Sheffield NHS Trust, Jessop Wing neonatal and obstetric departments, over a one year period. Analysis of the blood samples were conducted in the Academic Unit of Reproductive and Developmental medicine located at Jessop Wing, Royal Hallamshire Hospital. 8.2.2 Consent The Infants were recruited once informed consent was obtained from their parents/guardians. Infants whose parents refused consent were excluded from the study. 8.2.3 Inclusion Criteria All infants born at a gestational age greater than 24 weeks were eligible for the study. We chose to use the term ‘healthy’ to identify well from unwell preterm neonates who were deemed clinically stable by the attending clinical team. Preterm infants between 35-37 weeks admitted to the neonatal unit, transitional care ward or postnatal ward were establishing feeds or required prophylactic 85 Chapter 8 – Clinical Study A antibiotics pending blood cultures. Preterm infants less than 35 weeks were admitted to the neonatal unit primarily because of prematurity and low birth weight were generally well and establishing feeds. Extremely premature neonates (e.g. 24 weeks) who were ventilated or required continuous positive airway pressure (CPAP) for poor respiratory effort were clinically stable, and therefore considered to be ‘healthy’ for inclusion into the study. 8.2.4 Exclusion Criteria 1. The Infant born to mother whose pregnancy was complicated by diabetes (including gestational, insulin dependent and non-insulin dependent diabetes mellitus). 2. Deliveries associated with chorioamnionitis, or other significant antenatal or labour complications. 3. Infants with congenital and chromosomal abnormality. Rationale for exclusion criteria: Amylin has been shown to have both vasodilator (Hall and Brain 1999) and anti-inflammatory (Clementi, Caruso et al. 1995) properties. Since amylin shares 50% homology with calcitonin gene related peptide, which is recognised to be raised in acute inflammatory states such as trauma (Onuoha and Alpar 1999), sepsis (neonatal and adult) (Parida, Schneider et al. 1998), and hypotensive shock (Joyce, Fiscus et al. 1990), it may also share these properties. We therefore excluded infants born to mothers with chorioamnionitis and those with significant antenatal and labour complications. Similarly, amylin levels are influenced by diabetes and therefore mothers whose pregnancy was complicated by diabetes were also excluded from the study. 8.2.5 Study Procedure The research team approached the pregnant mothers for consent in the antenatal clinic and on admission to the labour ward prior to delivery. Mothers admitted to the labour ward for elective induction of labour and elective caesarean section were also approached. A pilot study highlighted concerns of consenting pregnant women during busy antenatal clinics and in active labour. Moreover it was acknowledged that pregnant mothers attending the antenatal 86 Chapter 8 – Clinical Study A clinics may not remember the information provided in the information leaflets at the time of admission to the labour ward. Therefore following feedback from mothers and midwives, pregnant women admitted for elective induction of labour and elective cesarean section were chosen as the target population. These pregnant mothers were identified by the research team from admission book and theatre lists in the antenatal ward and labour ward prior to elective induction of labour and elective caesarean section. The study was discussed with the prospective mothers and information leaflet provided. Written consent was obtained from mothers for collection of blood from umbilical cord and Guthrie bloods. Additionally, researchers also identified infants who were staying in the hospital for maternal reasons and who required Guthrie blood on day 5 and parents approached for consent. Blood was collected immediately after birth from umbilical cord stump that was clamped and separated from the baby. One to two ml of free flowing blood was collected from the umbilical vein and/or artery, were possible. The postnatal day 5 Guthrie blood was collected from in-house infants on the neonatal unit and postnatal wards and at home as a pilot for discharged babies. For in-house infants in the postnatal wards, special care baby unit and transitional care wards, 0.5-1.0 ml blood sample was collected from heel prick at the time of routine day 5 Guthrie. In infants admitted for neonatal intensive care, blood from capillary, vein or indwelling arterial catheters were obtained at the time of routine collection. Guthrie blood collection of term infants discharged home before their 5th day was conducted by their health visitors in the community. However community blood collection was abandoned due to the technical difficulty of maintaining cold chain and transporting the blood samples on ice. The study plan was therefore amended to recruit in-house infants alone who required Guthrie bloods during their stay in the neonatal intensive care, special care baby unit, transitional care and those staying in the hospital for maternal reasons. 8.2.6 Study End Point The study end point was reached once blood was collected at birth from the umbilical cord and postnatal period at the time of Guthrie test respectively. 87 Chapter 8 – Clinical Study A 8.2.7 Sample Collection & Storage Whole blood was drawn into refrigerated EDTA tube containing 500 IU of aprotinin, a serine protease inhibitor, known to inhibit trypsin, chymotrypsin, plasmin, kallikrein, and thrombin. Addition of aprotinin helps to inhibit protein degradation prior to separation of plasma (Mintz 1993) which was obtained by centrifugation immediately at 1000 xg for 10 minutes in refrigerated centrifuge. The plasma specimens were stored at ≤ -70°C. Blood samples with gross haemolysis or lipaemia were discarded as these were known to affect amylin levels. 8.2.8 Sample Analysis Linco research laboratories supplied two amylin assay kits namely, “Human amylin kit” and “Total amylin kit”. These amylin kits had been validated for use with human plasma and were sensitive to 1 pmol. The “Human amylin kit” reflected biologically active amylin more faithfully than “Total amylin kit”. In some cell types the reduced form (disulphide bond broken) is not biologically active although it may act as a competitive inhibitor (receptor antagonist) of the unreduced form. Since the disulphide bond is sometimes reduced in physiological conditions, the “Total amylin kit” tended to be higher than the “Human amylin kit”. The “Total amylin kit” was however considered to be more robust when reassurance about the controls with regards to the sampling variables, time to separation, and storage was not absolutely certain. This is because the rearrangements of the thiols in blood sample were common when there were unavoidable delays in processing due to strict clinical protocol. The research team concluded that it was impossible to strictly control sampling variables, time to separation and storage. Furthermore amylin assay developer and biochemical scientist opined that the marginal higher estimation of plasma amylin by “Total amylin kit” was very small to affect study results and conclusions. Therefore on balance, the “Total Human amylin kit” was chosen for the project. All plasma samples were analysed for amylin, and where volume of plasma was sufficient, insulin assay was also performed. Plasma total amylin immunoreactivity was quantified using Human Amylin (Total) ELISA KIT, a 88 Chapter 8 – Clinical Study A monoclonal antibody based sandwich immunoassay system developed by LINCO Research, Inc. (Missouri, USA). After the receipt of the ELISA kit, all components were stored at 2-8°C. This ELISA has sensitivity of 1 pmol (in 50 µl plasma) with an inter-assay coefficient of variation of 13-18% and intra-assay coefficient of variation of 2.5-5% at 50 pmol spiked concentration of amylin (Appendix 3). The basic steps of the assay are detailed in Appendix 2. Plasma immunoreactive insulin was measured by ELISA that has inter- and intra-assay coefficients of variation of 5.6% and 5.3% (BioSource Labs, Belgium) respectively. The principle of the sandwich ELISA (Appendix 1) is that the capture antibody and detection antibody binds to reduced or unreduced human amylin which is complexed with Streptavidin-Alkaline Phosphatase to form a sandwich. Substrate 4-Methylumbelliferyl Phosphate (MUP) is applied to the completed sandwich and fluorescent signal monitored at 355 nm/460 nm which is proportional to the amount of amylin present in the sample. 8.2.9 Sample Size A Priori statistical advice suggested a sample size of at least 200 observations from cord blood samples for the calculation of a reference interval (Altman DG 1991). 8.2.10 Statistical Analysis Statistical analysis was performed using Medcalc version 11.4.4.0. 1993-2010. Data were assessed for normality using coefficient of skewness, coefficient of kurtosis, D’Agostino-Pearson and Kolmogorov-Smirnov tests. Non-parametric tests (such as Wilcoxon rank sum test & Mann-Whitney U test) were employed where data were non-normal and presented as median values with interquartile range (25th-75th centile). The data were analysed for group differences with chisquare or Fisher’s exact tests for the categorical variables and with t-tests for the continuous variables. Pearson’s and Spearman’s correlation coefficient was used to assess correlation between normally and non-normally distributed amylin and insulin levels respectively. A p value of less than 0.05 was considered significant. A Bland-Altman plot of difference between the methods (y axis) against their average (x axis) was used to measure agreement between 89 Chapter 8 – Clinical Study A the two sampling methods (cord arterial and cord venous). This method aims to investigate a possible relation between measurement error and the true value which is unknown. Therefore the mean, average discrepancy (bias), trend, and scatter of the two measurements are used as a guide to the comparability of the two methods. 8.2.11 Data Collection & Case Report Form Case report forms (Appendix 14) of infants recruited into the study consisted of demographic details, clinical details, and time of blood sampling. Maternal details and clinical history were also entered on a CRF. 8.2.12 Study Summary Table 2 highlights the key features of the study. 90 Chapter 8 – Clinical Study A Table 2. Study A Summary Title Study Design Study Duration Objectives Number of blood samples Umbilical and Guthrie amylin levels in healthy preterm and term infants Single centre, Prospective Observational Study 12 months To determine plasma amylin levels in healthy preterm and term infants 200 umbilical cord samples / Participants Main Inclusion Criteria Infants ≥ 24 weeks gestation Infants Main Exclusion Criteria born pregnancy to was mothers whose complicated by diabetes Median and interquartile ranges for summary Coefficient Statistical Methodology statistics. to Corelation evaluate the relationship between variables. Bland Altman test for method comparison between umbilical umbilical venous arterial blood method Outcome variable Plasma amylin levels Measurement Assay ELISA Figure 19 below shows the overall study plan in a Gantt chart. 91 and sampling Chapter 8 – Clinical Study A Figure 19. Study A Gantt chart 8.3 Results Figure 20 below shows participant flow and collection of blood samples. 92 Chapter 8 – Clinical Study A 196 mothers eligible 14 mothers excluded due to significant perinatal complications 182 mothers consented 184 infants eligible 3 infants excluded (2 Downs syndrome, 1 CDH) Healthy Preterm infants (n=43) Healthy Term infants (n=138) Paired umbilical cord arterial blood where possible (n=26) Umbilical cord venous blood (n=138) Umbilical cord venous blood (n=43) Paired umbilical cord arterial blood where possible (n=8) 18 infants discharged on D1 124 infants discharged on D1 14 infants were inpatients for maternal reasons 25 infants were inpatient due to problems relating to prematurity (Post caesarean section) Guthrie Blood D5 (n=14) ELISA Guthrie Blood D5 (n=25) Cord Venous Amylin levels (181) Paired Cord Arterial Amylin levels (34) Figure 20. Study A flow diagram of participants and blood samples 93 Chapter 8 – Clinical Study A During the course of the study, 196 pregnant mothers (194 singleton and 2 twin gestation) were eligible for the study of whom 19 consented antenatally, 10 during early labour, and 169 prior to elective caesarean section and induction of labour. Fourteen mothers with singleton pregnancies subsequently developed severe perinatal complications (7 preeclampsia, 4 sepsis/chorioamnionitis, and 3 gestational diabetes requiring insulin therapy). Thus 182 mothers (180 singleton and 2 twin gestation) who consented gave birth to 184 infants. Three singleton mothers gave birth to infants who did not meet the inclusion criteria (2 Downs syndrome and 1 Congenital Diaphragmatic Hernia) and were therefore excluded from the study. Of the 181 infants who were eligible for blood collection, 138 were healthy term infants and 43 healthy preterm infants. Umbilical venous blood was collected from 138 term and 43 preterm infants. In addition, paired umbilical arterial blood was also collected from 26 term and 8 preterm infants. A total of 124 term and 18 preterm (> 35 weeks gestation) infants were discharged between D1-D4. Guthrie blood was collected from 14 term infants who were inpatient due to maternal reasons (post caesarean section) and 25 preterm infants who were inpatients due to problems associated with prematurity (9 feeding establishment, 5 temperature control, 7 mild respiratory distress, 4 monitoring for presumed sepsis). A total of 254 blood samples (181 umbilical cord venous, 34 paired (meaning when arterial blood was collected along with venous blood from the same umbilical cord where possible) umbilical cord arterial, and 39 Guthrie) were analysed for amylin levels. In addition, 33 paired umbilical cord blood samples were also analysed for insulin levels. The median (interquartile range) amylin concentration in 138 healthy term infants at birth (umbilical cord) was 6.10 (3.30-9.70) pmol/L. Table 3 shows the summary statistics of amylin concentration, birth weight and gestation at birth (umbilical cord group) in healthy term infants. Figure 21 provides the box and whisker plot showing distribution of individual amylin level at birth in 138 healthy term infants. 94 Chapter 8 – Clinical Study A Table 3. Summary statistic of amylin concentration and demographic characteristics at birth in healthy term infants Healthy Term Infants Birth weight Gestation Amylin (Umbilical Cord Group) (Kg) (Weeks) (pmol/L) Median 3.28 39.00 95% CI 3.15 – 3.42 38.00 - 39.00 5.20 - 7.49 25-75 percentile (IQR) 2.98 – 3.67 38.00 - 40.00 3.30 - 9.70 1.86-4.64 37-42 0.50-27.40 -0.06 0.60 (p=0.74) (p=0.00) 0.21 -0.43 (p=0.49) (p=0.26) 0.75 0.00 <0.00 Accept Reject Reject Min-Max Coefficient of Skewness Coefficient of Kurtosis *D-P test for Normal distribution * D’Agostino-Pearson test 95 6.10 1.14 (p<0.00) 1.49 (p=0.01) Chapter 8 – Clinical Study A 30 25 20 15 10 5 0 Healthy Term Infant Group (n=138) Figure 21. Box-and-whisker plot of umbilical cord amylin levels in healthy term infants The central box represents the values from 25th to 75th percentile, middle line represents the median, and the vertical line joining the horizontal line extends from the minimum to the maximum value (range), excluding "outside" values which are displayed as separate points. An outside value is defined as a value that is smaller than the lower quartile minus 1.5 times the interquartile range, or larger than the upper quartile plus 1.5 times the interquartile range. D’Agostino test for normal distribution was rejected (p<0.0001). The amylin concentration in 14 healthy term infants on postnatal day 5 (Guthrie) was 5.65 (3.10-8.20) pmol/L (Table 4 & Figure 22). 96 Chapter 8 – Clinical Study A Table 4. Summary statistic of amylin concentration and demographic characteristics at postnatal day 5 in healthy term infants Healthy Term Infants Birth weight Gestation Amylin (Guthrie Group) (Kg) (Weeks) (pmol/L) Median 3.40 38.5 95% CI 3.11-3.58 37.8-41.1 3.05-8.23 25-75 percentile (IQR) 3.12-3.58 38-41 3.10-8.20 Min-Max 2.02-4.42 37-42 1.4-21.0 -0.67 0.52 (p=0.24) (p=0.35) 3.23 -1.27 (p=0.05) (p=0.23) *DP test for Normal p=0.05 p=0.32 p=0.00 distribution Reject Accept Reject Coefficient of Skewness Coefficient of Kurtosis * D’Agostino-Pearson test 97 5.65 1.97 (p=0.00) 5.26 (p=0.00) Chapter 8 – Clinical Study A 25 20 15 10 5 0 Healthy Term Infant Guthrie Group (n=14) Figure 22. Box-and-whisker plot of Guthrie amylin levels in healthy term infants The central box represents 25th to 75th percentiles; middle line, median, and the vertical lines are limit lines (range) excluding "outside" value which is displayed as separate point. D’Agostino test for normal distribution was rejected (p value of p<0.003). There was no difference in the demographic characteristics and serum amylin levels at birth (umbilical cord) and postnatal day 5 (Guthrie) in healthy term infants (Table 5 & Figure 23). 98 Chapter 8 – Clinical Study A Table 5. Demographic characteristics of healthy term infants at birth and postnatal day 5 Healthy Term infants Birth (Umbilical Postnatal Day Cord venous) 5 (Guthrie) n 138 14 Amylin (pmol/L) 6.10 5.65 (3.30-9.70) (3.10-8.20) 39 38.5 (38.0-40.0) (38.0-41.0) 3.28 3.40 (2.98-3.67) (3.12-3.58) Male 71 7 Female 67 7 Gestation (wks) Birth Weight (kg) Gender * NS- not significant Median and interquartile range, Mann Whitney U test 99 p value *NS *NS *NS Chapter 8 – Clinical Study A 30 25 20 15 10 5 0 Umbilical Cord Group Healthy Term Infants (n=138) Guthrie Group Healthy Term Infants (n=14) Figure 23. Notched box-and-whisker plot of amylin levels at birth and postnatal day 5 in healthy term infants The central box represents 25th to 75th percentiles; the middle line, median and a vertical line, limits (range), excluding "outside" values which are displayed as separate points. The confidence intervals for the medians are provided by notches surrounding the medians which overlap; therefore the medians are not significantly different at a ± 95% confidence level. To test the difference between two groups, Mann- Whitney U test for independent samples was used, p=0.58. Table 6 shows the demographic characteristics in healthy preterm infants at birth. Although the group included preterm infants of 26 weeks gestation, the median gestational age at birth of 34 weeks suggests that the population studied were dominated by late preterm infants. The amylin level at birth (umbilical cord) in 43 healthy preterm infants was 4.60 (1.90-8.30) pmol/L (Table 6 & Figure 24). 100 Chapter 8 – Clinical Study A Table 6. Summary statistic of amylin concentration and demographic characteristics at birth in healthy preterm infants Healthy Preterm Infants Birth weight Gestation Amylin (Umbilical cord Group) (Kg) (Weeks) (pmol/L) Median 2.05 34.00 95% CI 1.65-2.30 33.00-34.39 3.16-6.19 25-75 percentile (IQR) 1.53-2.39 30.50-35.00 1.90-8.30 Min-Max 0.83-3.92 26-36 1.0-11.5 0.52 -0.87 0.35 (p=0.13) (p=0.02) 0.20 -0.36 (p=0.60) (p=0.47) p=0.29 p=0.05 Accepted Rejected Coefficient of Skewness Coefficient of Kurtosis *DP test for Normal distribution * D’Agostino-Pearson test 101 4.60 (p=0.31) -1.16 (p=0.12) p=0.18 Accepted Chapter 8 – Clinical Study A 12 10 8 6 4 2 0 Healthy Preterm Infant Group (n=43) Figure 24. Box-and-whisker plot of umbilical cord amylin levels at birth in healthy preterm infants The central box represents 25th to 75th percentiles; middle line, median, and the vertical lines, limit lines (range). D’Agostino test for normal distribution was accepted (p value of p=0.18). The amylin level on postnatal day 5 (Guthrie) was 6.9 (2.75-9.50) pmol/L (Table 7 & Figure 25) in 25 healthy preterm infants. 102 Chapter 8 – Clinical Study A Table 7. Summary statistic of amylin concentration and demographic characteristics at postnatal day 5 in healthy preterm infants Healthy Preterm Infants Birth weight Gestation Amylin (Guthrie Group) (Kg) (Weeks) (pmol/L) Median 1.72 33.0 95% CI 1.56-2.14 32.14-34.00 2.90-8.55 25-75 percentile (IQR) 1.50-2.24 30-35 2.75-9.50 Min-Max 0.83-2.75 26-36 1.0-19.7 0.00 -0.86 0.93 (p=0.99) (p=0.06) -0.62 -0.32 (p=0.37) (p=0.55) p=0.67 p=0.15 Accepted Accepted Coefficient of Skewness Coefficient of Kurtosis *DP test for Normal distribution * D’Agostino-Pearson test 103 6.9 (p=0.04) -0.08 (p=0.73) p=0.13 Accepted Chapter 8 – Clinical Study A 20 15 10 5 0 Healthy Preterm Infant Group (n=25) Figure 25. Box-and-whisker plot of amylin levels at postnatal day 5 in healthy preterm infants The central box represents the values from 25th to 75th percentile, middle line represents the median, and the horizontal line extends from the minimum to the maximum value, excluding "outside" value which is displayed as separate point. D’Agostino test for normal distribution was accepted (p value=0.13). There was no difference in the demographic characteristics and serum amylin levels of healthy preterm infants at birth (umbilical cord) and postnatal day 5 (Guthrie) (Table 8 & Figure 26). 104 Chapter 8 – Clinical Study A Table 8. Demographic charateristics of healthy preterm infants at birth and postnatal day 5. Healthy Preterm Birth (Umbilical Postnatal Day 5 Cord venous) (Guthrie) 43 25 Amylin (pmol/L) 4.60 (1.90-8.30) 6.9 (2.75-9.50) NS Gestation (wks) 34 (30.5 - 35.0) 33.0 (30.0 - 35.0) NS Birth Weight (kg) 2.05 (1.53 – 2.39) 1.72 (1.50 - 2.24) NS Male 25 13 Female 18 12 infants n Gender NS- not significant Median and interquartile range, Mann Whitney U test 105 P value Chapter 8 – Clinical Study A 20 15 10 5 0 Umbilical Cord Group Healthy Preterm Infants (n=43) Guthrie Group Healthy Preterm Infants (n=25) Figure 26. Notched box-and-Whisker plot of amylin levels at birth and postnatal day 5 in healthy preterm infants The central box represents 25th to 75th percentiles; the middle line, median; and vertical line the limits (range) excluding “outside" value which is displayed as a separate point. The confidence intervals for the medians are provided by notches surrounding the medians which overlap and therefore the medians are not significantly different at a ± 95% confidence level. Mann-Whitney U test for difference between the groups was not significant (p value=0.279). Independent sample t test was also not significant (p=0.07) The gestation and birth weight were significantly lower in preterm infants at birth (umbilical cord) and postnatal day 5 (Guthrie) compared to term infants. Amylin levels were significantly lower at birth (umbilical cord) but not on postnatal day 5 (Guthrie) in preterm infants when compared to term infants (Table 9 & Figure 27 & Figure 28). 106 Chapter 8 – Clinical Study A Table 9. Comparison of demography and amylin levels in healthy term and preterm infants at birth and postnatal day 5 Healthy Term Birth Healthy Preterm Birth Healthy Term Postnatal Day 5 Heathy Preterm Postnatal Day 5 138 43 14 25 Amylin (pmol/L) 6.10 (3.30 -9.70) *4.60 (1.90-8.30) 5.65 (3.10-8.20) **6.90 (2.759.50) *p=0.04 **p=0.63 Gestation (wks) 39 (38.0 - 40.0) *34 (30.5 - 35.0) 38.5 (38.0-41.0) **33.0 (30.0 - 35.0) *p<0.00 **p<0.00 Weight (kg) 3.28 (2.98-3.67) *2.05 (1.53- 2.39) 3.40 (3.12-3.58) **1.72 (1.50 - 2.24) *p<0.00 **p<0.00 Gender Male Female 71 67 25 18 7 7 13 12 n Median and interquartile range, Mann Whitney U test 107 p value Chapter 8 – Clinical Study A 30 25 20 15 10 5 0 Umbilical Cord Group Healthy Preterm Infants (n=43) Umbilical Cord Group Healthy Term Infants (n=138) Figure 27. Comparison Box-and-Whisker plot of amylin level in term and preterm infants at birth The central box represents 25th to 75th percentiles; the middle line, median; and vertical line, limits (range), excluding "outside" values. The confidence intervals for the medians are provided by means of notches surrounding the medians which do not overlap and therefore the medians are significantly different at a ± 95% confidence level, Mann-Whitney U test for independent samples was significant (p=0.04). 108 Chapter 8 – Clinical Study A 25 20 15 10 5 0 Guthrie Group Healthy Preterm Infants (n=25) Guthrie Group Healthy Term Infants (n=14) Figure 28. Comparison Box-and-Whisker plot of serum amylin level in term and preterm infants on postnatal day 5 The central box represents 25th to 75th percentiles; middle line, median; and vertical line, limits (range), excluding "outside" values. The notches about two medians overlap and therefore the medians are not significantly different at a ± 95% confidence level, Mann-Whitney U test for independent samples was not significant (p=0.93). Thus the study established reference interval for amylin concentrations at birth and postnatal day 5 in healthy term infants (6.10 (3.30-9.70) and 5.65 (3.108.20) pmol/L) and healthy preterm infants (4.60 (1.90-8.30) and 6.90 (2.759.50)) pmol/L respectively. 109 Chapter 8 – Clinical Study A Paired umbilical cord arterial and umbilical cord venous amylin levels were available from 34 infants. The median amylin level in 34 paired cord venous and arterial blood samples were 5.3 (2.97-10.17) and 5.8 (2.5-9.7) pmol/L respectively. The amylin levels in paired samples correlated positively (Spearman’s coefficient of rank correlation rho=0.94, 95% CI 0.89-0.97, p<0.0001) (Figure 29). 25 20 15 10 5 0 0 5 10 15 20 25 Umbilical Cord Venous Amylin Concentration (pmol/L) Figure 29. The correlation graph showing line of best fit to assess the degree of association between paired umbilical cord venous amylin (x axis) and umbilical cord arterial amylin (Y axis) Since the distribution of cord venous amylin levels were non-normal, rank correlation was used. The data were ranked in order of size, and calculations were based on the ranks of corresponding values X and Y to provide Spearman correlation coefficient rho with p-value, and a 95% CI for rho, i.e. the range of values which contains the true correlation coefficient with 95% probability (n=34, r=0.98, 95% CI 0.96-0.99, p<0.0001). 110 Chapter 8 – Clinical Study A A Bland-Altman plot (Figure 30 & Figure 31) or difference plot is a graphical method of the differences between cord venous and cord arterial amylin (y axis) against their average (x axis) to demonstrate agreement between the two sampling methods (cord venous versus cord arterial). 4 +1.96 SD 2.8 2 0 Mean -0.8 -2 -4 -1.96 SD -4.4 -6 -8 -10 0 5 10 15 20 25 AVERAGE of Cord Venous Amylin and Cord Arterial Amylin (pmol/L) Figure 30. Bland Altman method comparison plot of paired venous and arterial levels The plot is useful to reveal a relationship between the differences and the averages, to look for any systematic biases and to identify possible outliers. In this graphical method the differences between the two sampling techniques (Y axis) are plotted against the averages of the two techniques (X axis). Horizontal lines are drawn at the mean difference, and at the limits of agreement, which are defined as the mean difference plus and minus 1.96 times the standard deviation of the differences. Thus the average discrepancy between the two levels is -0.8 which is clinically small. The difference between the levels and scatter around the mean (bias) generally remain the same as the average increases (n=34, mean bias -0.8, and limits of agreement +2.8 to -4.4). 111 Chapter 8 – Clinical Study A 60 +1.96 SD 40 41.5 20 0 Mean -20 -13.9 -40 -60 -1.96 SD -80 -69.4 -100 -120 -140 0 5 10 15 20 25 AVERAGE of Cord Venous Amylin and Cord Arterial Amylin Figure 31. Bland Altman method comparison, percentage difference plot, of paired venous and arterial levels In this graphical method the differences are expressed as percentages of the values on the axis (i.e. proportionally to the magnitude of measurements). This option is useful when there is an increase in variability of the differences as the magnitude of the measurement increases. The Bland-Altman method aims to investigate a possible relation between measurement error and the true value which is unknown. Therefore the mean, average discrepancy (bias), trend, and scatter of the two measurements are used as a guide to the comparability of the two methods. The Bland-Altman plot is interpreted informally by investigating the size of the average discrepancy between the methods (bias), the trend of the the difference between methods (tend to get larger (or smaller) as the average increases), and the consistency of variability across the graph (Does the scatter around the bias line get larger as the average gets higher?). Horizontal lines are drawn at the mean difference, and at the limits of agreement, which are defined as the mean difference plus 112 Chapter 8 – Clinical Study A and minus 1.96 times the standard deviation of the differences. If the differences within mean ± 1.96 SD are not clinically important, the two methods may be used interchangeably. The mean bias was -0.8 and the limits of agreement +2.8 to -4.4 which is clinically acceptable. Thus umbilical venous or arterial blood may be used to analyse amylin levels in both term and preterm infants. Paired amylin and insulin levels were analysed from 33 umbilical cord blood samples from healthy preterm and term infants. The median insulin level at 38.5 (37-40) week gestation and birth weight 3.14 kg (2.61-3.76) was 5.0 (3.00-8.25) mu/L. The amylin levels showed statistical correlation with insulin levels (rho= 0.46, 95% CI=0.14-0.69, p<0.006) (Figure 32). However analysis of Figure 32 shows wide scatter suggesting this correlation is not strong. 113 Chapter 8 – Clinical Study A 25 20 15 10 5 0 0 5 10 15 20 Umbilical cord blood insulin concentration (mu/L) Figure 32. The correlation graph showing line of best fit to assess the degree of association between paired umbilical cord insulin (x axis) and umbilical cord amylin (Y axis) in healthy preterm and term infants Since the distribution of cord amylin (DP test, p=0.002) and insulin (DP test, p=0.0004) levels were non-normal, rank correlation was used. The data are ranked in order of size, and calculations were based on the ranks of corresponding values X and Y to provide Spearman correlation coefficient (rho) with p-value, and a 95% CI for rho, i.e. the range of values which contains the true correlation coefficient with 95% probability (n=33, spearman’s rho= 0.46, 95% CI=0.14-0.69, p<0.006). The umbilical cord venous amylin levels correlated both with gestation (Figure 33, r=0.20, 95% CI= 0.04-0.35, p=0.012) and birth weight (Figure 34, r=0.21, 95% CI=0.05-0.36, p=0.002). 114 Chapter 8 – Clinical Study A 30 25 20 15 10 5 0 25 30 35 40 45 Gestation at Birth (weeks) Figure 33. The correlation graph showing line of best fit to assess the degree of association between gestational age at birth (x axis) and umbilical cord amylin (Y axis) Since the distribution of cord venous amylin levels were non-normal, rank correlation was used to calculate Spearman correlation coefficient rho with pvalue, and a 95% CI for rho (n=181, Spearman’s rho=0.20, 95% CI= 0.04-0.35, p=0.012). 115 Chapter 8 – Clinical Study A 30 25 20 15 10 5 0 0 1000 2000 3000 4000 5000 Birth Weight (Grams) Figure 34. The correlation graph showing line of best fit to assess the degree of association between birth weight (x axis) and umbilical cord amylin (Y axis) Since the distribution of cord amylin levels were non-normal, rank correlation was used to calculate Spearman correlation coefficient rho with p-value, and a 95% CI for rho (n=181, Spearman’s rho= 0.21, 95% CI=0.05-0.36, p=0.002). 8.4 Discussion This single centre prospective observational study was the first to establish normal range of plasma amylin concentration in healthy term and preterm neonates at birth and at the fifth postnatal day. We chose to use the term ‘healthy’ to identify well from unwell preterm neonates who were deemed clinically stable by the attending clinical team. Term infants who stayed in the hospital for maternal reasons were well and did not need any medical interventions. Preterm infants between 35-37 weeks admitted to the neonatal unit, transitional care ward or postnatal ward were establishing feeds or required prophylactic antibiotics pending blood cultures. Preterm infants less 116 Chapter 8 – Clinical Study A than 35 weeks were admitted to the neonatal unit primarily because of prematurity and low birth weight were generally well and establishing feeds. Premature neonates who were ventilated or required continuous positive airway pressure (CPAP) for poor respiratory effort were clinically stable, and therefore included in the study. The plasma amylin levels at birth and postnatal day 5 in healthy term (6.10 and 5.65 pmol/L) and preterm infants (4.60 and 6.90 pmol/L) were similar to the levels observed (mean (SD)) in the paediatric (5.0 (1.94) pmol/l) and adult (5.0 (0.4) pmol/l) populations (Mitsukawa, Takemura et al. 1990; Akimoto, Nakazato et al. 1993). There was no difference in amylin concentration and birth weight, gestation, and gender between the umbilical cord and Guthrie group in both term and preterm infants. The amylin concentration in umbilical cord of preterm infants however was lower than terms infants (6.10 versus 4.60) most probably due to small sample size and differences in birth weight. In adults, like insulin, amylin levels show variation depending on fasting or postprandial state. It is not known if a similar pattern also exists in neonatal population. It is plausible that the amylin levels observed on postnatal day 5 in this study may have been influenced by the timing of blood sampling in relation to the infants’ pre-prandial or post-prandial state. This is less likely as the amylin levels were similar between the cord and Guthrie groups. We were unable to study diurnal variation of amylin and the influence of feeding due to the limitation of the study protocol to opportunistic blood collection. We propose that plasma amylin levels in the neonatal period are due to endogenous production as explained previously in section 2.4 on page 32, and are in the range reported in the paediatric and adult populations. Agreement was shown between paired cord arterial and venous amylin levels; therefore either of the two sampling methods could be used to measure amylin. This observation will help future studies investigating amylin levels in umbilical cord blood. Although umbilical cord amylin levels showed statistically significant correlation with insulin levels, due to wide scatter of the data, we suggest that amylin secretion may be in some way associated with insulin. In our study, umbilical 117 Chapter 8 – Clinical Study A vein insulin level from 34 neonates was 36.0 pmol/L (22.0-63.5) which was higher than 21.0 pmol/L (14.0-33.0) (Ong, Kratzsch et al. 2000) and in the range reported in previous studies (39.0±5 pmol/L and 33.0 pmol/L (24.0-41.0) (Thomas, de Gasparo et al. 1967; Gesteiro, Bastida et al. 2009). Fetal growth and development is dependent on various growth factors such as insulin, growth hormone and insulin like growth factor. The umbilical venous plasma insulin concentration and fetal insulin/glucose ratio increase exponentially with gestation, reflecting maturation of the endocrine pancreas (Economides, Nicolaides et al. 1990). Body weight at birth is related to the amount of functioning pancreatic tissue, with hyperinsulinemic babies being macrosomic and SGA babies having reduced amounts of pancreatic tissue (Fowden 1989). In this study, a positive correlation was observed between birth weight and umbilical vein insulin levels (Delmis, Drazancic et al. 1992). We also observed a direct correlation between amylin levels with ascending gestation and birth weight, finding shared with its cousin calcitonin gene related peptide (Parida, Schneider et al. 1998). It is possible that amylin may play a similar role to insulin in the maturation of pancreas. Although amylin levels showed an association with birth weight and gestation, the precise clinical significance of this observation is unclear. Since amylin is co-secreted with insulin from pancreas, we assessed cord amylin levels in a cohort of small for gestational age infants to assess the possible influence of faltering intrauterine growth. The median amylin level in the umbilical cord blood of 23 small for gestational age infants with median birth weight 1.80 kg (1.50-2.12) and gestation 36 weeks (33-37) was 4.0 (1.20-7.60) pmol/L. Although the amylin levels were lower that the levels observed in term and preterm infants, this did not reach statistical significance. Amylin has been shown to be raised in rat intestinal ischaemic injury (Phillips, Abu-Zidan et al. 2001), and may have a biologic role in sepsis (Parida, Schneider et al. 1998). Future research investigating the role of amylin in feed intolerance, necrotising enterocolitis and sepsis in the neonatal population may benefit from normal values established by this study. The main strength of the study was adherence to study methodology with regards to sample processing, storage and analysis. In addition, an experienced 118 Chapter 8 – Clinical Study A technician who was blinded to the patient details performed all laboratory analyses. To be absolutely certain that protein degradation does not take place in the blood collected prior to centrifugation, separation and storage, EDTA tubes were primed with aprotinin which is a protease inhibitor. The major limitations of the study were selection bias towards inpatient infants especially of term infants in the Guthrie group which also contributed to the small sample size; and blood sampling covering first postnatal week alone. Serial amylin measurements beyond first week of life in preterm and term infants may have helped understand the influence of transplacental passage, diurnal variations, influence of feeds and decay time of amylin. In hindsight, maternal amylin may have also helped determine transplacental passage of amylin. However we could not accommodate this into the study design due to logistical reasons. 8.5 Conclusions This single centre prospective observational study provides normative data of amylin levels in neonatal population. Further data from similar studies would be helpful in establishing a normal range. In healthy term infants, the median amylin level is 6.10 pmol/L at birth and 5.65 pmol/L at postnatal day 5. In healthy preterm infants, the median amylin level is 4.60 pmol/L at birth and 6.90 pmol/L at postnatal day 5. Umbilical cord venous amylin levels showed agreement with paired umbilical cord arterial amylin levels. Thus umbilical venous or arterial blood may be used to analyse amylin levels in term and preterm infants. The median insulin concentration in healthy preterm and term infants is 5.0 (3.00-8.25) mu/L and its correlation with amylin levels suggests an association and possibly co-secretion. Amylin levels demonstrate an association with gestation and birth weight, the exact significance of which is not clear. 119 Chapter 9 – Clinical Study B Chapter 9- Clinical Study B: Umbilical and Guthrie Amylin in Preterm and Term IMDM 9.1 Introduction In view of the physiological changes known to take place in fetus and neonate exposed to maternal diabetes, we hypothesised that amylin levels may be raised in new-born IMDM. The aim of this study was to determine serum amylin levels at birth (umbilical cord) and postnatal day 5 (Guthrie) in preterm and term IMDM. 9.2 Methodology 9.2.1 Study Design & Setting This single-centre prospective observational study was conducted at the University Hospitals of Sheffield NHS Trust neonatal and obstetric department, over a one year period. Analysis of the blood samples was conducted in the Academic Unit of Reproductive and Developmental medicine located at Jessop Wing, Royal Hallamshire Hospital. 9.2.2 Consent Infants were recruited once informed consent was obtained from their parents/guardians. Infants whose parents refused consent were excluded from the study. 9.2.3 Inclusion Criteria Infant’s ≥ 24 weeks gestation at birth born to mothers whose pregnancies were complicated by diabetes mellitus (including gestational, insulin dependent and non-insulin dependent diabetes mellitus) were eligible for the study. 9.2.4 Exclusion Criteria 1. Deliveries associated with chorioamnionitis, or other significant antenatal or labour complications. 2. Infants with congenital and chromosomal abnormality 120 Chapter 9 – Clinical Study B 9.2.5 Study Procedure Mothers whose pregnancies were complicated by diabetes were identified by the research team in the antenatal ward and labour ward before elective induction of labour and elective caesarean section. Information leaflet and consent were discussed with parents. Written consent was obtained for collection of blood from umbilical cord and during routine Guthrie. IMDM who stayed in the hospital for maternal reasons and requiring Guthrie bloods on day 5 were also identified. Additionally, IMDM admitted to the neonatal unit for monitoring of blood sugars were identified and parents approached for consent. Blood was collected immediately after birth from umbilical cord stump that was clamped and separated from the baby. One to two ml of free flowing blood was obtained from the umbilical vein and artery, where possible. Postnatal blood samples were collected at the time of routine day 5 Guthrie or during routine blood sampling (such as bedside glucose monitoring) from capillary, vein or indwelling arterial catheters as deemed necessary for routine collection. 9.2.6 Study End Point The infants were no longer followed up after collection of cord and postnatal bloods. 9.2.7 Sample Collection & Storage A 0.5 to 1 ml whole blood was collected and processed as described in Study A, section 8.2.7 on page 88. 9.2.8 Sample Analysis All samples were analysed for amylin, and where volume of plasma was sufficient, insulin assay was also carried out as described in study A, section 8.2.8 on page 88. 9.2.9 Sample Size At least 21 IMDM were required, to detect a difference in means of 15, assuming a standard deviation of 13 with 5% two sided significance level and 121 Chapter 9 – Clinical Study B 90% power. The mean and standard deviation were derived from the data published by (Mayer, Durward et al. 2002) 9.2.10 Statistical Analysis Statistical analysis was performed using Medcalc version 11.4.4.0. 1993-2010. as described in study A, section 8.2.10 on page 89. 9.2.11 Data Collection & Case Report Form Case report form (Appendix 15) of infants recruited into the study consisted of demography, clinical details, and time of blood sampling. Maternal details and clinical history were also entered on a CRF. 9.2.12 Study Summary Table 10 below highlights the key features of the study methodology. 122 Chapter 9 – Clinical Study B Table 10. Study B summary Title Umbilical and Guthrie amylin levels in preterm and term IMDM Study Design Prospective Observational Study Study Duration 12 months Study Centres Single Centre Objectives To determine plasma amylin levels in cord and Guthrie blood of IMDM infants Number of Samples/Participants 21 IMDM infants Main Inclusion Criteria IMDM ≥ 24 weeks gestation Statistical Methodology Median and interquartile ranges and non-parametric tests. Correlation Coefficient was performed to evaluate the relationship between variables. Outcome variable Plasma amylin levels Measurement Assay ELISA Figure 35 below provides details of the study plan in a Gantt chart. 123 Chapter 9 – Clinical Study B Figure 35. Study B Gantt chart 124 Chapter 9 – Clinical Study B 9.3 Result The Figure 36 below shows the participant flow and blood samples. 31 singleton mothers were eligible who consented Term infants (n=17) Umbilical cord venous blood Paired umbilical cord arterial blood (n=3) Guthrie blood (n=5) Feed intolerance (n=4) Preterm infants (n=14) Umbilical cord venous blood Paired umbilical cord arterial blood (n=3) Discharged on D1-4 (n=17) Inpatients Cord Amylin (n=31) Guthrie Amylin (n=14) Paired insulin (n=6) Guthrie blood (n=9) Feed intolerance (n=8) Figure 36. Study B-flow diagram of participants and blood samples Thirty-one singleton mothers whose pregnancy was complicated by diabetes consented for the study. 20 mothers had gestational diabetes and 11 mothers had insulin dependent diabetes mellitus. Umbilical cord blood was collected from 17 term infants and 14 preterm infants. Of these seventeen infants were discharged between days 1-4. Guthrie blood was collected from 5 term and 9 preterm inpatient infants who were admitted to the neonatal unit for respiratory distress, hypoglycemia, prematurity, or feeding problems. The null hypothesis of the study was that there was no difference in amylin concentration at birth and postnatal day 5 between preterm and term IMDM and preterm and term healthy infants (controls). The median (interquartile range) amylin concentration at birth (umbilical cord) in term IMDM was 34.30 (28.35-50.00) pmol/L (Table 11 & Figure 37). 125 Chapter 9 – Clinical Study B Table 11. Summary statistic of amylin concentration and demographic characteristics at birth in term IMDM infants Term IMDM Birth weight Gestation (umbilical cord Group) (kg) (weeks) Median 3.74 39.00 95% CI 3.44-3.98 38.00 - 39.98 32.79-47.39 3.41 – 4.07 37.75- 40.00 28.35-50.00 2.51-4.85 37-42 22.10-70.10 -0.25 0.58 (p=0.62) (p=0.26) -0.13 -0.21 (p=0.71) (p=0.66) 0.82 0.49 Accept Accept 25-75 percentile (IQR) Min-Max Coefficient of Skewness Coefficient of Kurtosis *D-P test for Normal distribution * D’Agostino-Pearson test 126 Amylin (pmol/L) 34.30 0.59 (p=0.25) -0.65 (p=0.417) 0.38 Accept Chapter 9 – Clinical Study B 80 70 60 50 40 30 20 IMDM Term Infant Umbilical Cord Group (n=17) Figure 37. Box-and-whisker plot of umbilical cord amylin levels in term IMDM infants The central box represents 25th to 75th percentile, middle line represents the median, and the vertical line represents range. D’Agostino-Pearson test for normal distribution of amylin levels was accepted (p=0.38). The plasma amylin level in term IMDM on postnatal day 5 (Guthrie) was 25.20 (22.20 – 48.75) pmol/L (Table 12 & Figure 38). 127 Chapter 9 – Clinical Study B Table 12. Summary statistic of amylin concentration and demographic characteristics on postnatal day 5 in term IMDM infants Term IMDM Birth weight Gestation (Guthrie Group) (kg) (weeks) Median 3.53 37 95% CI 2.67 – 4.67 35.22 – 40.77 12.18 – 56.29 25-75 percentile (IQR) 3.21 – 4.43 37 – 38.25 22.20 – 48.75 2.51-4.46 37-42 -0.54 2.23 0.85 -0.522 5.00 -0.988 (p=0.75) (p=0.02) Sample size too Sample size small too small Min-Max Coefficient of Skewness Amylin (pmol/L) 25.20 17.10-60.00 (p=0.74) Coefficient of Kurtosis *D-P test for Normal distribution (p=0.59) Sample size too small * D’Agostino-Pearson test Four of five infants in the Guthrie group also had feed intolerance at the time of blood collection. 128 Chapter 9 – Clinical Study B 60 55 50 45 40 35 30 25 20 15 IMDM Term Infant Guthrie group (n=5) Figure 38. Box-and-whisker plot of Guthrie amylin levels in term IMDM infants The central box represents the values from 25th to 75th percentile, middle line represents the median, and the vertical line joining the horizontal line extends from the minimum to the maximum value (range). The sample size was too small to test normal distribution. 129 Chapter 9 – Clinical Study B There was no difference in the demographic characteristics and plasma amylin levels of term IMDM at birth (umbilical cord) and postnatal day 5 (Guthrie) as shown in Table 13 & Figure 39. Table 13. Demographic characteristic of term IMDM at birth and postnatal day 5 Term IMDM Birth Postnatal Day (Umbilical Cord) 5 (Guthrie) 17 5 34.30 25.20 (28.35-50.00) (22.20-48.75) 39 37 (37.75-40.00) (37-38.25) 3.74 3.53 (3.41-4.07) (3.21-4.43) Male 8 2 *NS Female 9 3 *NS Group n Amylin (pmol/L) Gestation (wks) Weight (kg) p value *NS *NS *NS Gender * NS-not significant Median and interquartile range, Mann Whitney U test 130 Chapter 9 – Clinical Study B 80 70 60 50 40 30 20 10 Term IMDM Umbilical Cord Group (n=17) Term IMDM Guthrie Group (n=5) Figure 39. Comparison Box-and-Whisker plot of amylin levels at birth and postnatal day 5 in term IMDMs Mann-Whitney test for independent samples was not significant, p=0.25. 131 Chapter 9 – Clinical Study B In preterm IMDM, amylin level at birth (umbilical cord) was 32.0 (18.65-44.27) pmol/L (Table 14 & Figure 40). Table 14. Summary statistic of amylin concentration and demographic characteristics at birth in preterm IMDM infants Preterm IMDM Birth weight Gestation Amylin (umbilical cord group) (kg) (weeks) (pmol/L) Median 2.88 34 95% CI 1.97-3.51 33 – 35.46 18.51 –45.38 25-75 percentile (IQR) 1.97-3.35 33 – 35.25 18.65-44.27 Min-Max 0.82-4.21 26-36 13.90-90.00 -0.19 -1.69 1.25 (p=0.73) (p=0.00) -0.56 2.91 (p=0.49) (p=0.05) 0.74 0.00 <0.06 Accept Reject Accept Coefficient of Skewness Coefficient of Kurtosis *D-P test for Normal distribution * D’Agostino-Pearson test 132 32.00 (p<0.04) 1.28 (p=0.24) Chapter 9 – Clinical Study B 90 80 70 60 50 40 30 20 10 IMDM Preterm Infants Umbilical Cord Group (n=13) Figure 40. Box-and-whisker plot of umbilical cord amylin levels in preterm IMDM infants The central box represents 25th to 75th percentile, middle line represents the median, and the vertical line represents range. D’Agostino-Pearson test for normal distribution of amylin levels was accepted (p=0.06). 133 Chapter 9 – Clinical Study B In preterm IMDM, amylin level on postnatal day 5 (Guthrie) was 23.4 (15.3746.57) pmol/L (Table 15 & Figure 41). Table 15. Summary statistic of amylin concentration and demographic characteristics at postnatal day 5 in preterm IMDM infants Preterm IMDM Amylin Birth weight (pmol/L) (kg) (weeks) Median 23.4 2.61 34 95% CI 13.84 - 82.01 1.53 - 4.02 33.00 - 36.00 25-75 percentile (IQR) 15.37 - 46.57 1.96 - 3.92 38.0 - 40.0 4.90-92.00 0.82-4.21 26-36 1.38 -0.05 -1.45 (p<0.05) (p=0.92) 0.38 -1.22 (p=0.64) (p=0.30) 0.14 0.59 Accept Accept (Guthrie group) Min-Max Coefficient of Skewness Coefficient of Kurtosis *D-P test for Normal distribution * D’Agostino-Pearson test 134 Gestation (p=0.03) 1.58 (p=0.22) 0.05 Accept Chapter 9 – Clinical Study B 100 90 80 70 60 50 40 30 20 10 0 IMDM Preterm Infants Guthrie Group (n=9) Figure 41. Box-and-whisker plot of Guthrie amylin levels in preterm IMDM infants The central box represents 25th to 75 percentile, middle line represents the median, and the vertical line represents range. D’Agostino-Pearson test for normal distribution of amylin levels was accepted (p=0.14). At the time of Guthrie blood collection, eight of nine preterm IMDM were experiencing feed intolerance. 135 Chapter 9 – Clinical Study B There was no difference in the demographic characteristics and amylin levels of preterm IMDM at birth (umbilical cord) and postnatal day 5 (Guthrie) as shown in Table 16 & Figure 42. Table 16. Demographic characteristics of preterm IMDM at birth and postnatal day 5 Preterm IMDM n Amylin (pmol/L) Gestation (wks) Weight (kg) Birth Postnatal day 5 (Umbilical cord) (Guthrie) 14 9 32.0 23.4 (18.65-44.27) (15.37-46.57) 34 34 (33-35.25) (38-40) 2.88 2.61 (1.93-3.35) (1.96-3.92) Gender P value *NS *NS *NS *NS Male 9 5 Female 5 4 * NS-not significant Median and interquartile range, Mann Whitney U test 136 Chapter 9 – Clinical Study B 100 90 80 70 60 50 40 30 20 10 0 Umbilical Cord Group Preterm IMDM (n=13) Guthrie Group Preterm IMDM (n=9) Figure 42. Comparison box-and-whisker plot of amylin concentration at birth and postnatal day 5 in preterm IMDM Mann-Whitney test for independent samples did not reach statistical significance, p=0.48. 137 Chapter 9 – Clinical Study B Figure 43 below summarises the serum amylin levels at birth and postnatal day 5 in term and preterm IMDM. There was no statistical difference in plasma amylin levels between term and preterm IMDM at birth (p=0.25) and postnatal day 5 (p=0.64). 100 90 80 70 60 50 40 30 20 10 0 TermIMDMCord PretermIMDMCord TermIMDMGuthrie PretermIMDMGuthrie p=0.25 Figure 43. p=0.64 Comparison box-and-whisker plot of amylin concentration in umbilical cord and Guthrie blood in term and preterm IMDM Mann-Whitney test for independent samples was not significant, cord group p=0.25 and Guthrie group p=0.64. 138 Chapter 9 – Clinical Study B There was no baseline difference in the demographic characteristics at birth (umbilical cord) and postnatal day 5 (Guthrie) between term IMDM and term healthy controls (Table 17). However, the median amylin level at birth in term IMDM was significantly increased at 34.30 pmol/L compared to 6.10 pmol/L in term healthy controls (p <0.0001). Similarly, the median amylin level at postnatal day 5 in term IMDM was also significantly increased at 25.20 pmol/L compared to 5.65 pmol/L in term healthy controls (p < 0.0001) (Figure 44). Table 17. Demographic characteristics and serum amylin levels of term healthy controls and term IMDM at birth and postnatal day 5 n Amylin (pmol/L) Gestation (wks) Term Controls Birth Term IMDM Birth Term Controls Postnatal Day 5 Term IMDM Postnatal Day 5 138 17 14 5 5.65 (3.10-8.20) **25.20 (22.20-48.75) 38.5 (38.00-41.00) 37.0 (37-38.25) NS NS 6.10 (3.30-9.70) *34.30 (28.35-50.00) 39.00 39 (38.00-40.00) (37.75-40.00) Weight (kg) 3.28 (2.98-3.67) 3.74 (3.41-4.07) 3.40 (3.12-3.58) 3.53 (3.21-4.43) Gender Male Female 71 67 8 9 7 7 2 3 NS-not significant Median and interquartile range, Mann Whitney U test 139 p value *p<0.00 **p<0.00 Chapter 9 – Clinical Study B 80 70 60 50 40 30 20 10 0 HealthyTermCord TermIMDMCord p<0.0001 HealthyTermGuthrie TermIMDMGuthrie P<0.001 Figure 44. Comparison box-and-whisker plot of amylin levels at birth and postnatal day 5 between term IMDM and healthy term infants (controls) Mann-Whitney test for independent samples was significant, cord group p<0.0001 and Guthrie group p <0.001. The null hypothesis was therefore rejected. Thus amylin levels are significantly raised in term IMDM both at birth and postnatal day 5 when compared to term healthy control infants. 140 Chapter 9 – Clinical Study B There was no difference in the demographic characteristics at birth (umbilical cord) and postnatal day 5 (Guthrie) in preterm IMDM and preterm healthy controls (Table 18). However, the median amylin level at birth in preterm IMDM was significantly increased at 32.00 pmol/L compared to 4.60 pmol/L in preterm healthy controls (p<0.0001). Similarly, the median amylin level at postnatal day 5 in preterm IMDM was also significantly increased at 23.40 pmol/L compared to 6.90 pmol/L in preterm healthy controls (p <0.0001) (Figure 45). Table 18. Demographic characteristics and serum amylin levels of preterm healthy controls and preterm IMDM at birth and postnatal day 5 Preterm Controls Birth Preterm IMDM Birth Preterm Controls Postnatal Day 5 Preterm IMDM Postnatal Day 5 43 14 25 9 4.60 (1.90-8.30) *32.00 (18.65-44.27) 6.90 (2.75-9.50) **23.40 (15.37-46.57) Gestation 34 (wks) (30.5 - 35.0) 34 (33-35.25) 33 (30.0 - 35.0) 34 (38-40) NS Weight (kg) 2.88 (1.97-3.35) 1.72 (1.50 - 2.29) 2.61 (1.96-3.92) NS n Amylin (pmol/L) Gender Male Female 2.05 (1.53-2.39) p value *p<0.00 **p<0.00 NS 25 18 9 5 13 12 NS-not significant Median and interquartile range, Mann Whitney U test 141 5 4 Chapter 9 – Clinical Study B 90 80 70 60 50 40 30 20 10 0 HealthyPretermCord PretermIMDMCord HealthyPretermGuthrie TermIMDMGuthrie p<0.0001 P<0.0001 Figure 45. Comparison box-and-whisker plot of amylin levels at birth and postnatal day 5 between preterm IMDM and healthy preterm infants (controls) Mann-Whitney test for independent samples was significant, cord group p <0.0001 and Guthrie group p<0.0001. The null hypothesis was therefore rejected. Thus serum amylin levels are significantly raised in preterm IMDM both at birth and postnatal day 5 when compared to healthy preterm infants (controls). Paired amylin and insulin levels were analysed from 3 IMDM preterm and 3 IMDM term infants from umbilical cord blood samples. The median insulin level was 10.5 (8.0-16.5) mu/L. Increased positive correlation was observed when this data were combined with paired amylin and insulin levels from healthy preterm and term infants (n=39, Spearman’s rho= 0.56, 95% CI=0.29-0.74, p<0.0001) (Figure 46). 142 Chapter 9 – Clinical Study B 100 80 60 40 20 0 0 5 10 15 20 25 Umbilical Cord Insulin Concentration (mu/L) Figure 46. The correlation graph showing line of best fit to assess the degree of association between umbilical cord insulin (x axis) and umbilical cord amylin (Y axis) from paired umbilical cord blood samples of healthy and IMDM preterm and term infants Since the distribution of cord venous amylin levels were non-normal, data were ranked in order of size, and calculations were based on the ranks of corresponding values X and Y to provide Spearman correlation coefficient rho with p-value, and a 95% CI for rho, (n=39, Spearman’s rho= 0.56, 95% CI=0.290.74, p<0.0001). Thus the median insulin level in preterm and term IMDM was twice (n=6, 10.5 (8.0-16.5) mu/L) the level observed in healthy preterm and term infants (n=33, 5.0 (3.00-8.25) mu/L) and its correlation with amylin levels confirms an association and possibly co-secretion. 143 Chapter 9 – Clinical Study B 9.4 Discussion This single centre prospective observational study was the first to determine plasma amylin concentrations in term and preterm IMDM at birth and fifth postnatal day. An alternative to observational methodology would have been case-control study, IMDM with feed intolerance (cases) compared to IMDM without feed intolerance (controls). Case-control study incorporating objective assessment of gastric emptying in the study methodology may have added validity to the study results. However not all infants with feed intolerance were admitted to the neonatal unit or necessitated introduction of oro/nasogastric tube, a prerequisite to identify infants with feed intolerance. Furthermore, the issues of funding, technical expertise and ethical concerns of performing gastric emptying tests in neonatal setup were considered as major limiting factors. Interestingly, 4/5 term infants who required admission to the neonatal unit for monitoring of blood sugars were also found to have feed intolerance with increased prefeed gastric residual volumes and frequent vomiting. 8/9 preterm infants were also experiencing feed intolerance at the time of Guthrie blood collection. Due to small number of the admitted infants who were also experiencing feed intolerance we could not draw meaningful relationship of amylin levels to the degree of feed intolerance or resolution of symptoms of feed intolerance. In addition we could not identify any obvious differences in the characteristics of IMDM who experienced feed intolerance with those who did not. It is possible that minor but important differences in relation to the degree of feed tolerance and amylin levels were missed as we did not set out to study objective measures of feed intolerance. Further studies are warranted to shed more light on this topic. In this study serum amylin levels obtained from cord and Guthrie blood were significantly raised in infants born to mothers whose pregnancy was complicated by diabetes and these levels were comparable to those reported in critically ill children with feed intolerance (Mayer, Durward et al. 2002). We suggest that these high serum amylin levels observed in IMDM may be partially responsible for the feed intolerance seen in this population. This is in keeping with the physiological action of amylin, as it is 15–20 times more potent than 144 Chapter 9 – Clinical Study B other known inhibitors of gastric motility (Young 1997). There are a number of possible explanations for such high levels observed in IMDM. Amylin is deficient in type 1 diabetes mellitus and is present in excess in conditions in which insulin is hyper-secreted (Rink, Beaumont et al. 1993). It would therefore seem plausible that transient neonatal hyperinsulinaemia in IMDM as a consequence of in-utero hyperglycaemia, may also stimulate hypersecretion of endogenous amylin. This is further supported by evidence of co-secretion of amylin and insulin, shown by the positive correlation between amylin and insulin in study A and Study B (n=39, spearman’s rho= 0.56, 95% CI=0.29-0.74, p<0.0001) and previous studies (Rink, Beaumont et al. 1993; Mayer, Durward et al. 2002). Moreover, the median insulin level in preterm and term IMDM was twice (n=6, 10.5 (8.0-16.5) mu/L) the level observed in healthy preterm and term infants (n=33, 5.0 (3.00-8.25) mu/L) suggesting possible fetal and neonatal hyperinsulinaemia. Unfortunately we were unable to obtain sufficient sample volumes for analysis of paired amylin and insulin on the fifth postnatal day. It is plausible that the amylin levels observed on postnatal day 5 in preterm and term IMDM in this study may have been influenced by the timing of blood sampling in relation to the infants’ pre-prandial or post-prandial state. This is less likely as the amylin levels were similar between the cord and Guthrie groups in both preterm and term IMDM. We were unable to study diurnal variation of amylin and the influence of feeding due to the limitation of the study protocol to opportunistic blood collection. Although, the median amylin levels on the fifth postnatal day were lower than umbilical cord samples at birth in both preterm and term IMDM, they did not reach statistical significance. This is most likely to be due to small sample size and comparatively lower gestation and birth weight possibly due to a selection bias towards blood collection of inpatient neonates in the Guthrie group. However a more plausible explanation for this gradual decline in Guthrie amylin levels in preterm and term IMDM is possibly explained by declining hyperinsulinaemic environment. This would also coincide with the time course of resolution of the feed intolerance observed in 4 term IMDM infants who were discharged by day 7 established to full feeds. We did not set out to study these infants in more detail and therefore cannot draw any meaningful conclusions about specific factors relevant to the group of infants who displayed feed 145 Chapter 9 – Clinical Study B intolerance. The observed serum amylin levels in preterm and term IMDMs on the fifth postnatal day (23.4 and 25.20 pmol/L) were still above the normal range observed in healthy preterm and term infants (6.90 and 5.65 pmol/L). The high amylin levels in IMDM further support the theory that transplacental passage either does not occur or is minimal as serum amylin levels are subnormal in diabetic patients (Ludvik, Lell et al. 1991; Sanke, Hanabusa et al. 1991; Koda, Fineman et al. 1992; Akimoto, Nakazato et al. 1993). Thus the observed increased amylin levels in IMDM would seem to represent endogenous amylin production. AC187 is a truncated amylin peptide antagonist that acts by selectively and potently blocking amylin receptors (Young, Gedulin et al. 1994). The biological actions of amylin in IMDM need to be elucidated prior to evaluation of pharmacological blockade of this peptide as a potential therapeutic option. The strength of the study was strict adherence to study protocol with regards to sample processing and storage. However, a major limitation of the study was selection bias to inpatient IMDM which also contributed to small numbers especially in the Guthrie group. Although it was possible to collect serial blood for amylin levels beyond first week of postnatal life, we did not include this in our methodology fearing refusal by the ethics committee. Inclusion in the study methodology of maternal amylin concentration and serial amylin measurements in preterm and term IMDM may have helped understand the influence of transplacental passage, diurnal variations, possible influence of feeds and decay time of amylin. Moreover, this study did not attempt to correlate amylin levels with objective measures of gastric emptying and feed intolerance or its resolution. This is an important limitation of the study and may be considered in the design of future studies. In conclusion, we have presented evidence in support of our hypothesis that amylin levels are raised in preterm and term IMDM which may contribute to the observed feed intolerance. 146 Chapter 9 – Clinical Study B 9.5 Conclusions The amylin levels in term and preterm IMDM at birth and postnatal day 5 were significantly higher than levels observed in healthy preterm and term controls. Amylin by virtue of its potent inhibition of gastric emptying may play a role in the pathogenesis of feed intolerance in this group of infants. The cord insulin levels in preterm and term IMDM were twice the levels observed in the cord blood from healthy preterm and term infants. These levels correlated positively with amylin levels which suggest an association with insulin secretion. 147 Chapter 10 – Clinical Study C Chapter 10- Clinical Study C: Amylin Levels in Preterm infants with Feed Intolerance 10.1 Introduction Amylin is a potent inhibitor of gastric emptying and its elevated levels are shown to be associated with delayed gastric emptying in critically ill children with feed intolerance (Mayer, Durward et al. 2002). We hypothesized that preterm neonates with feed intolerance evidenced by increased prefeed GRV (surrogate marker for delayed gastric emptying) may have high plasma amylin concentrations. The main objective of this study was to determine serum amylin concentrations in preterm neonates with and without feed intolerance. 10.2 Methodology 10.2.1 Study Design & Setting This two-centre case-control study was conducted at the University Hospitals of Sheffield NHS Trust and Doncaster General Hospital neonatal departments, over a one year period. Analyses of the blood samples were conducted in the Academic Unit of Reproductive and Developmental medicine located at Jessop Wing, Royal Hallamshire Hospital. 10.2.2 Consent Infants were recruited once informed consent was obtained from their parents/guardians. Infants whose parents refused consent were excluded from the study. 10.2.3 Inclusion Criteria 1. Preterm infants between 24-36 weeks gestation at birth admitted to the neonatal intensive care unit and requiring gastric tube placement and 2. Preterm infants who were receiving > 30 ml/kg or > 25% of total fluid intake as enteral feeds through a gastric tube. 148 Chapter 10 – Clinical Study C 10.2.4 Exclusion Criteria 1. Deliveries associated with intrauterine growth retardation, abnormal antenatal Doppler assessment (risk factors for NEC which may influence amylin levels), maternal infection, chorioamnionitis, pre-eclampsia and significant labour complications (maternal inflammatory conditions which may influence amylin levels). 2. Infants born to mothers whose pregnancy were complicated by diabetes mellitus (due to conclusions of Study B). 3. Infants with congenital abnormalities, chromosomal abnormalities (possible bias due to association with feed intolerance), structural gastrointestinal abnormalities, necrotising enterocolitis, and sepsis (possible bias due to association with feed intolerance and in addition may influence amylin levels). 4. Infants requiring administration of prokinetic agents including erythromycin. 5. Exclusive breast feeding infants in who prefeed gastric residual volume cannot be determined. 6. Haemoglobin less than 7 g/dl (due avoid further blood loss). Rationale for study exclusion criteria: Amylin is raised in IMDM as shown in study B, acute inflammatory conditions, maternal and neonatal sepsis, and in rat intestinal ischaemic injury (possible role in necrotising enterocolitis). It shares 50 % homology with CGRP which has anti-inflammatory and vasodilator function and is also shown to be raised in patients with sepsis and soft tissue injury (Joyce, Fiscus et al. 1990; Clementi, Caruso et al. 1995; Parida, Schneider et al. 1998; Hall and Brain 1999; Onuoha and Alpar 1999; Phillips, Abu-Zidan et al. 2001; Kairamkonda, Deorukhkar et al. 2005). This data from animal and human studies of conditions which may influence amylin concentrations informed the study exclusion criteria. 10.2.5 Study Procedure The feeding regime of the neonates was dictated by the local guideline. Expressed breast milk (EBM) was encouraged, and formula feed was used if breast milk was not available. The neonates were fed every hour and the 149 Chapter 10 – Clinical Study C volumes increased as clinically tolerated but not more than 20mls/kg birth weight/day. Every four hours pre-feed GRV were recorded as a percentage of the previous feed volume. Abdominal girth, colour of gastric residuals and emesis were also noted. If this GRV was greater than 50% on two consecutive occasions, neonates were classified as feed-intolerant (nTOL). The further management of these neonates was up to the attending neonatologist. Researcher or responsible consultant identified preterm infants who were receiving > 30 ml/kg or > 25% of total fluid intake as enteral feeds through a gastric tube. Parents were approached for consent and information leaflets provided. In neonates with feed intolerance (nTOL), 0.5-1 ml blood was collected from capillary, vein, or indwelling arterial catheters as deemed necessary for routine collection; within 2 hours of the second big (> 50%) prefeed GRV. Feed-tolerant (TOL) neonates served as controls. The researchers identified TOL infants matched for gestational age and birth weight and wherever possible postnatal age with nTOL infants. 0.5-1 ml blood was collected from these neonates at the time of next routine collection. All blood handling was performed as per guidance for the minimization of infection risk to both participants and investigators. 10.2.6 Definition of feed intolerance Infants whose Gastric Residual Volume (GRV) was greater than 50% on two consecutive occasions were identified as feed-intolerant (nTOL). We chose this definition of feed intolerance partly due to personal and published observation by Mayer et al (Mayer, Durward et al. 2002) who defined critically ill children as feed-intolerant on the basis of GRV greater than 125% 4 hour after a feed challenge which correlated with three objective measures of gastric emptying. In addition, based on the assumption that stomach empty’s every 2 hours, we concluded that in infants in our study who were fed every hour, GRV of more than 50% on 2 consecutive occasions would improve the sensitivity of the definition. This definition was different from previously reported in that the target GRV had to be proved on 2 consecutive occasions which we felt would improve the accuracy and sensitivity to the definition. It was also acknowledged that the proportion of feed that may be aspirated is strongly influenced by the size of the feed and hence the inclusion criteria was limited to those preterm infants who were receiving > 30 ml/kg or > 25% of total fluid intake as enteral feeds. Thus 150 Chapter 10 – Clinical Study C the minimal feed volume at any given time was more than 2mls even in the smallest infant. 10.2.7 Definition of feed tolerance Infants with minimal (prefeed GRV <50%) or no GRV for > 24 hours were identified as feed tolerant (TOL). 10.2.8 Study End Point The infants were no longer followed up after collection of bloods. 10.2.9 Sample Collection & Storage A 0.5 to 1 ml whole blood was drawn into ice-cooled EDTA tube containing 500 IU of aprotinin. The separation of plasma and storage was performed as explained in Study A, section 8.2.7 on page 88. 10.2.10 Sample Analysis All samples were analysed as described previously in section 8.2.8 on page 88. 10.2.11 Sample Size To detect a minimum difference of 15 pmol/L between the feed tolerant (TOL) and intolerant (nTOL) groups, with a power of 90%, assuming a standard deviation of 13, a total number of 20 neonates per group had to be recruited for significance levels of 5% (a=0.05). The mean and standard deviation for the sample size calculation was based on data from previous studies in neonates and children (Mayer, Durward et al. 2002; Kairamkonda, Deorukhkar et al. 2005) 10.2.12 Statistical Analysis Statistical analysis was performed using Medcalc version 11.4.4.0. 1993-2010. Individual group data were assessed for normal distribution by applying coefficient of Skewness and Kurtosis and D’Agostino-Pearson test. The coefficient of Skewness and Kurtosis is a measure for the degree of symmetry and peakedness/flatness respectively in the variable distribution. The D'Agostino-Pearson test (Sheskin 2003) computes a single p-value for the 151 Chapter 10 – Clinical Study C combination of the coefficients of Skewness and Kurtosis. The tests for normal distribution were accepted when the p value was > 0.05. Normal data were analysed for group differences with chi-square or Fisher’s exact tests for the categorical variables and with t-tests for the continuous variables. Median and interquartile range (25th–75th centile) and non-parametric tests such as Wilcoxon rank sum test & Mann-Whitney U test were employed where data were non-normal. Pearson’s correlation coefficient (r) or Spearman’s rank test (rho) was used to assess correlation between variables with parametric and non-parametric distribution respectively. A p value of less than 0.05 was considered significant. 10.2.13 Data Collection & Case Report Form The Case report form (Appendix 16) of infants recruited into the study consisted of demography, clinical details, and time of blood sampling. Maternal details and clinical history were also entered on a CRF. 10.2.14 Study Summary Table 19 & Figure 47 below highlight the key features of the study methodology and Gantt chart respectively. 152 Chapter 10 – Clinical Study C Table 19. Study C summary Title Amylin levels in preterm infants with feed intolerance Study Methodology Case-Control Study Study Duration 12 months Study Centres Two Centre Objectives To determine plasma amylin levels in preterm infants with feed intolerance Number of Participants 20 nTOL and 20 TOL Main Inclusion Criteria Preterm infants (24-36 weeks) admitted to the neonatal intensive care unit and requiring gastric tube feeding Statistical Methodology Median and interquartile ranges and non-parametric tests. Coefficient be will Correlation performed to evaluate the relationship between variables Outcome variable Plasma amylin levels Measurement Assay ELISA 153 Chapter 10 – Clinical Study C Figure 47. Study C Gantt chart 154 Chapter 10 – Clinical Study C 10.3 Results Figure 48 below shows study participant flow and timing of blood collection. 70 preterm infants were eligible for the study once they met the selection criteria. Blood was collected from 34 feed intolerant (nTOL) and 36 feed tolerant (TOL) infants. 4 blood samples in the nTOL and 6 in the TOL group were heavily hemolysed and therefore discarded. Thirty blood samples in each group were available for further analysis. Consent obtained from parents of preterm neonates (24-36 weeks) with gastric tube placement n=70 Enteral feeds >30ml/kg or >25% of total fluid intake >50% prefeed GRV on two consecutive occasions None or <50% prefeed GRV Study entry and consent Study entry and consent Feed-intolerant (nTOL) (n=34) Feed-tolerant (TOL) (n=36) n=6 excluded Blood samples haemolysed & therefore unsuitable for analysis n=4 excluded Blood samples haemolysed & therefore unsuitable for analysis Feed-tolerant (TOL) (n=30) Feed-intolerant (nTOL) (n=30) Figure 48. Study C-Flow diagram of participants and blood samples The null hypothesis of the study was that serum amylin levels in preterm infants with feed intolerance were no different to preterm infants without feed intolerance. 155 Chapter 10 – Clinical Study C There was no significant difference between the nTOL and TOL group with regards to baseline characteristics such as gestation, birth weight, gender, postnatal age, ventilation days, CPAP days, type of feed, blood glucose, white cell count and CRP (Table 20). Table 20. Baseline characteristics at the time of blood sampling of preterm neonates defined as feed-intolerant (nTOL) and feed-tolerant (TOL) nTOL (n=34) TOL (n=36) p value n Gestation (wks) 34 29.5 (28-31) 36 30.0 (29-33) 0.33 Birth weight (kg) 1.30 (1.00-1.80) 1.30 (1.10-1.80) 0.62 Gender Male (n) Female (n) 23 11 24 12 0.64 0.62 7.5 (4-10) 9.5 (5-12) 0.16 Ventilation (days) 1 (0-4) <1 (0-3) 0.21 CPAP (days) 3 (0-4) 2 (0-5) 0.41 22 7 5 24 7 5 0.12 0.26 0.18 Blood glucose (mmol/l) 4.2 (3.5-4.9) 3.9 (3.1-5.0) 0.23 White cell count (109/l) 9.0 (7.5-10.4) 8.3 (6.3-10.4) 0.08 6 (6-10) 6 (6-10) 0.51 Postnatal age (days) Feed type EBM (n) Formula (n) Mixed (n) C reactive protein (mg/l) Data are presented as median (interquartile range) Amylin concentration, % GRV and time to full enteral feeds were significantly raised in nTOL compared to TOL group. However, there was no difference in length of hospital stay between nTOL and TOL group (Table 21). 156 Chapter 10 – Clinical Study C Table 21. Serum amylin levels, % GRV, days to reach full feeds and length of stay in feed-intolerant (nTOL) and feed-tolerant (TOL) neonates n Amylin (pmol/L) GRV (%) Time to full feeds (days) Length of hospital stay (days) nTOL TOL P value 30 30 47.9 (21.4-79.8) 8.7 (5.7-16) <0.00 150 (100-350) 5 (0-5) <0.00 13.0 (10.0-16.0) 6.5 (4.0-9.0) <0.00 52.5 (21-69) 37 (20-50) 0.08 Data are presented as median [interquartile range] GRV (%)-percentage gastric residual volume prior to next scheduled feed measured prior to amylin sampling In particular, median amylin level was significantly raised in nTOL group at 47.9 pmol/L compared to 8.7 pmol/L in TOL group. The null hypothesis was therefore rejected. Thus amylin levels in preterm infants with feed intolerance are significantly raised compared to preterm infants without feed intolerance. Figure 49, Figure 50 & Figure 51 show box and whisker plots of plasma amylin, % GRV and time to reach full enteral feeds in nTOL and TOL group. 157 Chapter 10 – Clinical Study C 120 100 80 60 40 20 0 nTOL (n=30) TOL (n=30) Figure 49. Box and whisker plot of amylin levels (y-axis) in feed-intolerant (nTOL) and feed-tolerant (TOL) neonates (x-axis) D’Agostino-Pearson test for normal distribution for amylin levels was accepted for nTOL (p=0.21) and rejected for TOL (p<0.0001) group. The central box represents 25th to 75th percentile, middle line represents the median, and the vertical line, range, excluding "far out" values (red circles). Median amylin level was significantly raised in nTOL compared to TOL group (47.9 vs 8.7) (MannWhitney test for independent samples was significant, p value <0.0001). The details of the 2 outliers in the TOL group are as follows. The male infant with amylin level 76.1 pmol/L was born at 29+5 weeks gestation with birth weight 1.44 kg to a 33 year mother without any medical problems. He was fed expressed breast milk and was on caffeine at the time of blood collection on day 12. The male infant with amylin level 67.7 pmol/L was born at 29 weeks gestation with birth weight 1.14 kg to a 26 year mother who presented to labour ward with premature rupture of membranes for > 12 hours and abnormal cardiotocograph. The infant had initial features of hypoxic ischaemic encephalopathy which settled in three days. He was also treated for coagulase negative staphylococcal infection and abdominal distension with feed 158 Chapter 10 – Clinical Study C intolerance until day 12. He was fed nutriprem milk with occasional vomiting and was on caffeine at the time of blood collection on day 28. 450 400 350 300 250 200 150 100 50 0 TOL (n=30) nTOL (n=30) Figure 50. Box and whisker plot of percentage GRV (y axis) in feed-intolerant (nTOL) and feed-tolerant (TOL) neonates (x axis) D’Agostino-Pearson test for normal distribution was accepted for nTOL (p=0.105) and rejected for TOL (p<0.0001). The central box represents 25th to 75th percentile, middle line represents the median, and the vertical line represents range, excluding "outside" (black circles) and "far out" (red circles) values which are displayed as separate points. %GRV was significantly raised in nTOL compared to TOL (150% vs 5%) (Mann-Whitney test for independent samples was significant, p value <0.0001). 159 Chapter 10 – Clinical Study C 50 40 30 20 10 0 nTOL (n=30) TOL (n=30) Figure 51. The box and whisker plot of days to reach full feeds (Y axis) in feedintolerant (nTOL) and feed-tolerant (TOL) neonates (X axis) D’Agostino-Pearson test for normal distribution was rejected for nTOL and TOL (p<0.001). The central box represents 25th to 75th percentile, middle line represents the median, and the vertical line represents range, excluding "outside" (black circles) and "far out" (red circles) values which are displayed as separate points. Days to reach full enteral feeds was significantly increased in nTOL compared to TOL group (13 days vs 5.5 days) (Mann-Whitney test for independent samples was significant, p value <0.0001). 160 Chapter 10 – Clinical Study C In the nTOL group, plasma amylin levels correlated with percentage gastric residual volumes (Figure 52), days to reach full enteral feed volume of 150 ml/kg/day (Figure 53) and days to discharge (Figure 54). 120 nTOL Group (n=30) 100 80 60 40 20 0 0 100 200 300 400 500 Percentage Gastric Residual Volume (%GRV) nTOL Group (n=30) Figure 52. Correlation graph showing line of best fit to assess the degree of association between amylin concentration (y axis) and % GRV (x axis) in feedintolerant (nTOL) group D’ Agostino-Pearson test for normal distribution was accepted for both % GRV (p=0.10) and amylin levels (p=0.21). n=30, Pearson correlation coefficient, r=0.78, p<0.0001, 95%CI 0.59-0.89. 161 Chapter 10 – Clinical Study C 120 nTOL Group (n=30) 100 80 60 40 20 0 5 10 15 20 25 30 35 Days To Full Enteral Feeds nTOL Group (n=30) Figure 53. The correlation graph showing line of best fit to assess the degree of association between amylin concentration (y axis) and days to reach full enteral feeds (x axis) in nTOL group D’ Agostino-Pearson test for normal distribution was rejected for days to reach full enteral feeds (p=0.001) and accepted for amylin levels (0.21). n=30, Spearmans coefficient of rank correlation rho=0.56, p=0.001, 95% CI 0.26-0.77. 162 Chapter 10 – Clinical Study C 120 nTOL Group (n=30) 100 80 60 40 20 0 0 20 40 60 80 100 120 Days To Discharge nTOL Group (n=30) Figure 54. The correlation graph showing line of best fit to assess the degree of association between days to discharge (x axis) and amylin concentration (Y axis) in the feed-intolerant (nTOL) group D’ Agostino-Pearson test for normal distribution was accepted for days to discharge (p=0.49) and amylin levels (0.21). n=30, Pearson’s correlation coefficient r=0.43, p=0.015, 95%CI 0.092-0.68. 163 Chapter 10 – Clinical Study C In the TOL group, plasma amylin levels did not correlate with percentage gastric residual volumes (Figure 55), days to reach full enteral feed volume of 150 ml/kg/day (Figure 56), and days to discharge (Figure 57). 80 70 TOL Group (n=30) 60 50 40 30 20 10 0 0 5 10 15 20 25 Percentage Gastric Residual Volume (%GRV) TOL Group (n=30) Figure 55. The correlation graph showing line of best fit to assess the degree of association between % GRV (x axis) and amylin levels (y axis) in feed-tolerant (TOL) group D’Agostino-Pearson test for normal distribution was rejected for % GRV and amylin (p<0.001). n=30, Spearman’s coefficient of rank correlation rho=0.33, p=0.068, CI -0.025-0.622. 164 Chapter 10 – Clinical Study C 80 70 TOL Group (n=30) 60 50 40 30 20 10 0 0 10 20 30 40 50 Days To Full Enteral Feeds TOL Group (n=30) Figure 56. The correlation graph showing line of best fit to assess the degree of association between days to full enteral feeds (x axis) and amylin levels (Y axis) in feed-tolerant (TOL) group D’Agostino-Pearson test for normal distribution was rejected for days to reach full enteral feeds and amylin (p<0.001). n=30, Spearman’s coefficient of rank correlation rho=-0.003, p=0.98, CI -0.363-0.357. 165 Chapter 10 – Clinical Study C 80 70 TOL Group (n=30) 60 50 40 30 20 10 0 0 20 40 60 80 100 120 Days To Discharge TOL Group (n=30) Figure 57. The correlation graph showing line of best fit to assess the degree of association between days to discharge (x axis) and amylin levels (Y axis) in the feed-tolerant (TOL) group D’Agostino-Pearson test for normal distribution was accepted for days to discharge (p=0.11) and rejected for amylin (p<0.001). n=30, Spearman’s coefficient of rank correlation rho=-0.33, p=0.07, -0.61-0.03. In conclusion the study results confirm the hypothesis that plasma amylin levels are raised in preterm infants with feed intolerance. 10.4 Discussion Up to 67% preterm very low birth infants struggle to attain full enteral nutrition in the first few weeks of life (Reddy, Deorari et al. 2000; ElHennawy, Sparks et al. 2003; Patole, Rao et al. 2005; Hay 2008). Feed intolerance is the common cause of this problem and is characterised by large gastric residual volumes (GRV) with or without associated clinical manifestations (Jadcherla and Kliegman 2002; Mihatsch, von Schoenaich et al. 2002; Patole 2005). Reliance on total parenteral nutrition during this period is usually not enough to maintain protein and lipid intake (Grover, Khashu et al. 2008; Hay 2008) and this often 166 Chapter 10 – Clinical Study C leads to problems with infection and hepatic damage (Donnell, Taylor et al. 2002; Kaufman, Gondolesi et al. 2003). Notably these infants may lag behind in protein and calorie intake by up to 6-8 weeks by the time they are ready to go home (Grover, Khashu et al. 2008) which may have adverse effects on their postnatal growth and neurodevelopment (Frank and Sosenko 1988; Hack, Breslau et al. 1991; Ehrenkranz, Younes et al. 1999; Hack, Schluchter et al. 2003; Cooke, Ainsworth et al. 2004; Hay 2008). Delayed gastric emptying, intestinal immaturity, ileus of prematurity, and gastro-esophageal reflux may all play a role. It is not clear whether one or all of these mechanisms contribute to the observed feed intolerance. This is the first study to evaluate the role of amylin as an inhibitor of gastric motility in feed intolerance in the neonatal intensive care setting. The results of this study support the hypothesis that plasma amylin levels are increased in neonates with delayed gastric emptying compared to controls and these levels correlate with increasing GRV. We propose that although poor gastric motility in premature neonates is probably multifactorial in origin, amylin may play a role in its pathophysiology. The amylin levels in nTOL were approximately 3.5 times that of TOL controls. These levels were comparable to those reported in critically ill children with feed intolerance (Mayer, Durward et al. 2002) and are amongst the highest reported levels in humans. This is in keeping with the physiological action of amylin, as it is 15-20 times more potent than other known inhibitors of gastric motility (Young 1997). The amylin levels in the TOL group (8.7 pmol/L) were comparable to the Guthrie levels in healthy preterm neonates (6.90 pmol/L). Although the median postnatal timing (9.5 days) of amylin sampling was delayed in TOL controls, this difference was not significant compared to nTOL group (7.5 days). The main reason for this delay was because the routine collection of blood in TOL controls was less frequent. However the blood samples from nTOL and TOL were treated in the same way and therefore did not bias the observed results. The proposed mechanism of action of amylin is outlined below in Figure 58. It is plausible that amylin may play a similar role on gastric emptying in preterm neonates with feed intolerance. 167 Chapter 10 – Clinical Study C Figure 58. Proposed pathway of amylin action Nutrient intake into stomach not only stimulates insulin release but also amylin release which inhibits gastric emptying through brain centres regulating gastric motility. Amylin also inhibits glucagon release and further food intake, thus reducing endogenous glucose production in the postprandial period. The reason for increased amylin levels in nTOL neonates is unclear. We could only speculate the possible reasons for this hyperamylinemia in the nTOL group. As amylin is co-secreted with insulin under normal physiological conditions, an attractive hypothesis is that transient hyperinsulinemia and relative insulin resistance in nTOL infants may stimulate amylin release. This is based on the observation that many extremely preterm infants develop hyperglycemia in the first week of life during continuous glucose infusion due to relative insulin resistance and defective islet beta cell processing of proinsulin (Mitanchez-Mokhtari, Lahlou et al. 2004). Furthermore compared with term 168 Chapter 10 – Clinical Study C neonates; proinsulin and C-peptide levels were higher in normoglycemic preterm infants less than 30 weeks (23.2 +/- 0.9 vs 18.9 +/- 2.71 pmol/L and 1.67 +/- 0.3 vs 0.62 +/- 0.12 nmol/L, respectively). In addition, among low birth weight infants, fat mobilization and beta oxidation of fatty acids in the liver is not suppressed by glucose stimulated insulin secretion suggesting decreased insulin sensitivity (Hemachandra and Cowett 1999). Interestingly the associated blood glucose abnormalities were not observed in the study infants and furthermore the glucose levels were comparable between nTOL and TOL group. We speculated that the reason for this observation was because insulin secretion in neonates is less closely linked to circulating blood glucose concentrations (Hawdon, Aynsley-Green et al. 1993; Mitanchez-Mokhtari, Lahlou et al. 2004). However we could not speculate the reasons for the absence of hyperinsulenemia and insulin resistance in the TOL infants. It is known that large intravenous dextrose loads may promote amylin release (Mitsukawa, Takemura et al. 1990); however the study neonate’s received standard glucose maintenance and remained euglycemic. Although the major site of amylin biosynthesis is within pancreatic islet β cells, amylin secretion other than nutrient mediated stimulation cannot be ruled out. In addition, other potential physiological mediators of gastric emptying including GLP-1 and CCK may potentiate the actions of amylin (Young 1997). The effects of possible interaction of amylin with other gut peptides remain to be studied in preterm population. Transplacental passage giving rise to raised amylin levels in nTOL group seems unlikely as discussed previously in section 2.4 on page 32. Studies A and B and a previous study (Mayer, Durward et al. 2002) showed correlation between serum amylin and insulin levels indicating a degree of preservation of pancreatic hormonal co-release. Although ideally we would like to have repeated this in this study we were limited ethically by the blood volume we could take in very low birth weight neonates. Unfortunately we were also unable to measure insulin resistance. It is plausible that the amylin levels observed in nTOL and TOL group in this study may have been influenced by the blood sampling time and relationship to the infants’ prandial state. This is less likely due to the observations highlighted in section heading 2.2 and previous study observations as highlighted in section heading 8.4 and 9.4. Notably twenty-three of thirty nTOL bloods were collected 169 Chapter 10 – Clinical Study C when the infants were nil by mouth and 7/30 before the next scheduled feed (reduced or same volume). Thus the observed amylin levels in nTOL could be considered as baseline and pre-prandial in the nTOL group. We did not observe an association of amylin levels to the timing of blood collection in the TOL group probably due to the short interval between scheduled feeds (1-2 hours). The mean stomach half emptying time in infants fed expressed breast milk was shorter than formula fed infants (Ewer, Durbin et al. 1994). Additionally, gastric distension (which may be influenced by bolus or continuous milk feeds) may stimulate amylin release and resultant slowing of gastric emptying (Reda, Geliebter et al. 2002). The types of milk feed were comparable between the groups and the neonates in both the study groups received intermittent bolus feeds. The percentage GRV is influenced by the absolute feed volume and feeding interval. It could be argued that the difference in % GRV simply reflects much smaller absolute feed volume in the nTOL group and therefore the amount of aspirate would be a much greater percentage. We do not think that the absolute feed volumes or intervals influenced % GRV observed between the groups as only 3 infants in the nTOL group had absolute feed volume less than 2 mls/hr. Amylin levels correlated with percentage GRV and time to reach full enteral feeds which were significantly increased in the nTOL group. Although it is conceivable that gestational age or birth weight might have been responsible for this association, a similar observation was not demonstrated in the TOL group. Furthermore both gestational age at birth and birth weight were not statistically different between nTOL and TOL groups. This has clinical significance since increased length of time to reach full enteral feeding is associated with a poorer neurological outcome (Patole 2005). Amylin may thus be used as an early marker of delayed enteral nutrition. Although the length of stay was increased in nTOL compared to TOL group, it did not reach statistical significance even after stratification by gestation and birth weight. Studies investigating feed intolerance in preterm population could be facilitated by the availability of a consistently reliable clinical or biochemical marker of feed intolerance. Percentage prefeed GRV to define feed intolerance has been 170 Chapter 10 – Clinical Study C validated in children and adults against more objective measures of gastric emptying including paracetamol absorption test, ultrasonography and scintigraphic techniques (Mayer, Durward et al. 2002; ElHennawy, Sparks et al. 2003; Gomes, Hornoy et al. 2003; Pozler, Neumann et al. 2003). We formulated a pragmatic definition of feed intolerance based on consensus opinion, physiology of gastric emptying in preterm infants and previously published definitions. Our definition of feed intolerance is similar to those used in previous trials (Ng, So et al. 2001; Salhotra and Ramji 2004). Also the plasma amylin levels correlated with % GRV in the nTOL group adding validity to the definition of feed intolerance in this study. Thus more than 50% prefeed GRV may be used as a clinical indicator of delayed gastric emptying. With the prevailing fear of NEC and its association with feed intolerance most caregivers are often persuaded to investigate colour of prefeed GRV, abdominal distension, vomiting, the presence of blood in stool and apnoea and bradycardia. However the clinical significance of each of these criteria to establish feeding strategies and for feeding intolerance has yet to be determined. Most clinicians and nurses determine daily feeding strategy based on greenish aspirates, vomiting or abdominal girth. We also monitored and recorded abdominal girth, colour of the gastric residuals and emesis. Notably green gastric residuals in ELBW infants were not negatively correlated with feeding volume on day 14 (Mihatsch, von Schoenaich et al. 2002). Only seven infants in this study showed vomiting and abdominal distension (more than 2cm) at the time of sample collection (4 nTOL and 3 TOL). Although this is a small sample size, we did not see an association with NEC, a delayed time to achieve full enteral feeds, PDA or amylin levels. Amylin is raised in IMDM, and may be affected by intestinal ischaemic injury and acute inflammatory conditions (Joyce, Fiscus et al. 1990; Clementi, Caruso et al. 1995; Parida, Schneider et al. 1998; Hall and Brain 1999; Onuoha and Alpar 1999; Phillips, Abu-Zidan et al. 2001; Kairamkonda, Deorukhkar et al. 2005) influencing our exclusion criteria. The inflammatory markers such as Creactive protein, white cell count and neutrophil count were comparable between the groups and therefore not attributable to the observed difference in the amylin levels. 171 Chapter 10 – Clinical Study C Amylin was found to be significantly elevated in the ischaemia-reperfusion group that had clamping of the superior mesenteric artery for 60 min followed by 15 min reperfusion (median 39 pM, range 30-44) compared with the Sham operated group that underwent laparotomy and isolation (without clamping) of the superior mesenteric artery (19 pM, range 15-45; Mann-Whitney U, p < 0.05) and the Control group that underwent no surgery (24 pM, range 15-55; p < 0. 01) (Phillips, Abu-Zidan et al. 1999). The same researchers observed a positive correlation between the histologic score of the intestinal injury and the measured plasma amylin concentration (R = 0.48, P =.007) (Phillips, Abu-Zidan et al. 2001). Thus amylin is increased in intestinal ischeamia and its levels are related to the degree of intestinal ischaemic injury in rats. However none of the infants in nTOL and TOL groups experienced necrotising enterocolitis or significant abdominal pathology. In addition, amylin levels were not associated with vomiting and abdominal distension (more than 2cm) in 4 nTOL and 3 TOL infants at the time of sample collection. Objective assessment of gastric emptying may have added validity to this study, however with intra-observer variability, safety, ethical issues, and the need for repeated blood sampling in our preterm population render these assessments prohibitive. Serial measurement of amylin levels may have helped to establish the critical amylin level associated with resolution of feed intolerance. However multiple samples could not be taken from this population. A degree of selection bias was unavoidable, as we aimed to include neonates with and without gastric stasis, reflective of our intensive care population. The strengths of the study were the adherence to sample processing and storage protocol and the blood samples were obtained during times when blood was collected for routine clinical monitoring which minimised any extra discomfort to the infant. This study has implications for future research in preterm infants with feed intolerance. In particular, further studies are warranted to establish beyond doubt that amylin is responsible for delayed gastric emptying in preterm neonates. Furthermore, although we established that neonates with feed intolerance have high amylin levels; it is unclear why amylin is increased in these preterm but well neonates. The possible areas of interest are the role of hyperinsulinemia, insulin resistance, amylin receptor insensitivity and other gut peptides. This study may also stimulate future research into the effect of bolus 172 Chapter 10 – Clinical Study C versus continuous feeds on amylin secretion (stretch receptors) and gastric emptying in preterm population. Amylin receptor antagonists are being developed for animal studies. It would be premature to discuss pharmacological blockade of amylin peptide as one of the therapeutic option in feed intolerance until further studies firmly establish the conclusions of this study. In addition considerable caution is warranted as it is not known if amylin by virtue of its association with feed intolerance in preterm infants plays a protective role on the immature gut. In addition there is a possibility of adverse effects due to the blockade of amylin’s other biological functions. 10.5 Conclusions Amylin by virtue of its inhibitory action on gastric emptying may play a role in its etiology. The study results confirmed the hypothesis that serum amylin levels are increased in preterm infants with feed intolerance compared to preterm infants without feed intolerance. Serum amylin concentration, percentage gastric residual volume and time to reach full enteral feeds were significantly raised in nTOL group compared to TOL group. Thus measurement of serum amylin level in preterm infants may have clinical and revenue implications. Definition of feed intolerance in preterm infants (> than 50% prefeed GRV prior to the next scheduled feed) correlated with amylin levels. Increased GRV may be associated with feed intolerance and may be a useful surrogate marker for feed intolerance in preterm neonates. 173 Chapter 11 – Conclusions of the dissertation Chapter 11- Conclusions of the Dissertation Amylin is a potent inhibitor of gastric emptying and plays an important role in glucose homeostasis. Feed intolerance is common in preterm infants and infants born to mothers whose pregnancy was complicated by diabetes. We therefore set out to test the hypothesis that serum amylin levels are increased in preterm infants with feed intolerance. We also hypothesized that serum amylin levels are raised in infants born to mothers whose pregnancy was complicated by diabetes. It was evident from literature that there was no uniform validated definition of feed intolerance in neonatal population. Additionally a gold standard test for gastric emptying in new-born population especially preterm infants was not available. Therefore a pragmatic definition of feed intolerance was formulated based on consensus opinion, physiology of gastric emptying in preterm infants and previous published literature. The primary aims of the study were to (i) establish normal serum amylin concentrations at birth and postnatal day 5 in healthy preterm and term infants (ii) determine amylin levels in infants of diabetic mothers and (iii) determine amylin levels in preterm infants with feed intolerance. The secondary aims were to (i) investigate if amylin and insulin were indeed co-secreted (ii) investigate agreement of serum amylin levels in paired umbilical cord arterial and venous blood (iii) investigate the relationship between serum amylin concentrations and gastric residual volumes (GRV) (iv) investigate the relationship between serum amylin concentrations and length of hospital stay. To meet the aims and objectives, three studies were designed and conducted in parallel. Prospective observational and case control study designs were adopted to answer the hypotheses. The median serum amylin levels in term infants at birth and postnatal day 5 were 6.10 (3.30-9.70) and 5.65 (3.10-8.20) pmol/L respectively. The median serum amylin in preterm infants at birth and postnatal day 5 were 4.60 (1.90174 Chapter 11 – Conclusions of the dissertation 8.30) and 6.90 (2.75-9.50) pmol/L respectively. These levels were comparable to those seen in children and adults. Amylin like insulin may have ontogenetic significance as it correlated with both birth weight and gestation. Paired amylin levels agreed in umbilical cord arterial and venous blood. Either sampling method could therefore be used in future research involving amylin levels in cord bloods. Amylin and insulin levels correlated in paired blood samples suggesting an association between these peptides and possible co-secretion. Amylin levels were significantly increased at birth and postnatal day 5 in preterm (32.00 and 23.40) and term (34.30 and 25.20) infants born to mothers whose pregnancy was complicated by diabetes which may explain the observed feed intolerance. Amylin levels were significantly raised in preterm neonates with delayed gastric emptying compared to controls (47.9 vs 8.7 pmol/L). The serum amylin levels correlated with gastric residual volumes providing validity of GRV as a marker of feed intolerance due to delayed gastric emptying. Thus the study definition of feed intolerance (prefeed residual gastric volumes of more than 50% on 2 consecutive occasions) may be considered to be reliable for future studies. The serum amylin levels in preterm neonates also correlated with time to reach full enteral feeds and days to discharge which has clinical and revenue implications. Overall the study has achieved its intended aims and objectives. The study made original contribution to establishing normative data of amylin levels in the new-born population and explored its significance in infants born to mothers whose pregnancy was complicated by diabetes and preterm infants with feed intolerance. 175 Chapter 12 – Implications for future research Chapter 12- Implications for Future Research There are several areas that I was able to identify during the course of this study that have implications for future research. One of the major handicaps to analytical studies in preterm population is the volume of blood required for ELISA and similar tests. Our study methodology was therefore restricted to opportunistic blood collections due to the volume of blood required especially in preterm infants. Dried blood spot (DBS) technology has been shown to be useful in pharmacokinetic-pharmacodynamic (PK-PD) studies including preterm population. Although this technology is expensive, future study designs limited by volume of blood collected in preterm infants should explore this technique. It is important to establish placental passage of amylin to make sense of the postnatal amylin concentrations we established in new-born population. Future studies of this hormone should investigate maternal amylin levels prior to birth of their baby. This is especially relevant in infants of diabetic mothers in addition to measurement of amylin levels from umbilical cord blood and serial postnatal samples. Alternatively studies on perfused human placenta at various amylin concentrations may help answer this question conclusively. Amylin is increased in rat intestinal ischaemia (Phillips, Abu-Zidan et al. 2001) and may have a biologic role in sepsis (Parida, Schneider et al. 1998). Future research exploring the role of amylin in necrotising enterocolitis and sepsis in the neonatal population may benefit from normal values established by our study. It is theorised that exposure to maternal diabetes and alterations in fetal glucose, insulin, or other factors results in altered fetal adaptation and changes in normal fetal programming (Pettitt, Aleck et al. 1988). Evidence of altered fetal programming (reduced insulin sensitivity) is shown to be present before birth in large for gestation neonates (Dyer, Rosenfeld et al. 2007). Thus high amylin levels in IMDM could be explained by hyperinsulinaemia. However the reason for increased amylin levels in feed intolerant preterm neonates is unclear. As amylin is co-secreted with insulin under normal physiological conditions, an attractive hypothesis that could be put to clinical test is that transient 176 Chapter 12 – Implications for future research hyperinsulinemia and relative insulin resistance may stimulate amylin release. The adult gold standard of euglycemic clamp to study insulin resistance would not be feasible and safe in neonatal population due to the invasive nature of the procedure. However it is possible to study glucose/insulin and insulin/cortisol ratios (Gesteiro, Bastida et al. 2009) to assess insulin resistance in neonatal population. Other possible areas of interest are the role of amylin receptor insensitivity and interaction with other gut peptides such as CCK, GLP-1, leptin and ghrelin. Other potential physiological mediators of gastric emptying including GLP-1 and CCK may potentiate the actions of amylin (Young 1997). Therefore studies investigating the interaction with other gut peptides including ghrelin may help understand the conundrum of the pathophysiology of gastric emptying in newborn infants with feed intolerance. There is an urgent need for validation of noninvasive techniques for gastric emptying assessments in new-born infants and preterm extremely low birth weight infants. Employment of these validated tests of gastric emptying in future studies may add validity to the biological role of amylin and other gut peptides in the pathophysiology of feed intolerance in humans. Amylin release is also known to be influenced by gastric distension and stretch receptors. The results of this study may stimulate future research into the effect of bolus versus continuous feeds on amylin secretion (stretch receptors) and gastric emptying in preterm population. Amylin receptor antagonists are being developed for research purposes in animals. Amylin receptor blockade would seem to be one of the potential therapeutic options for feed intolerance in preterm infants. However considerable caution is warranted as it is not known if amylin by causing feed intolerance in preterm neonates has a protective role on the immature gastrointestinal tract. As highlighted before amylin has other important biological function in addition to gastric emptying and glucose homeostasis. Thus pharmacological blockade of this peptide as a potential therapeutic option could expose the preterm infant to greater risk of necrotising enterocolitis in addition to other possible adverse effects. 177 Chapter 12 – Implications for future research Obesity is a worldwide problem with significant health and economic problems. Amylin by virtue of its central and peripheral action on the stomach acts as an anorectic hormone. Thus synthetic analogues of amylin may be developed as anorectic agents that may have therapeutic implications in the management of childhood and adult obesity. 178 Chapter 14 – Appendices Chapter 13- Publications arising from this Project 13.1 Amylin peptide levels are raised in infants of diabetic mothers. Arch Dis Child. 2005 Dec; 90(12):1279-82 179 Chapter 14 – Appendices 180 Chapter 14 – Appendices 181 Chapter 14 – Appendices 182 Chapter 14 – Appendices 183 Chapter 14 – Appendices 13.2 Amylin peptide is increased in preterm neonates with feed intolerance. Arch Dis Child Fetal Neonatal Ed. 2008 Jul; 93(4):F265-70 184 Chapter 14 – Appendices 185 Chapter 14 – Appendices 186 Chapter 14 – Appendices 187 Chapter 14 – Appendices 188 Chapter 14 – Appendices 189 Chapter 14 – Appendices Chapter 14- Appendices Appendix 1.Basic principle of luminescent ELISA The basic steps in any luminescent ELISA are as follows: 1. Antibodies or antigens are passively adsorbed to solid surfaces such as commercially available 96-well format microtiter plates for use in ELISA. 2. As one of the reactants in the ELISA is attached to a solid-phase, the separation of bound and free reagents is made by simple washing procedures. 3. The result of an ELISA is a color reaction that can be observed by eye and read rapidly using specially designed multichannel spectrophotometers allowing data to be stored and analysed statistically (Crowther 1995) . Analyte Analytespecific antibody Step 1. Analyte-specific antibody (capture antibody) is pre-coated onto a microplate. The sample is added and any analyte present is bound by the immobilized antibody. 190 Chapter 14 – Appendices AntibodyAnalyte Conjugate Step 2. An enzyme-linked analyte-specific detection antibody binds to a second epitope on the analyte forming the analyte-antibody complex. Substrate- Light or Colour Step 3. Substrate is added and converted by the enzyme, thereby producing a colored product or light emission in direct proportion to the amount of analyte bound in the intial reaction (signal increases as concentration increases). Step 4. A standard curve is prepared from the data produced from the serial dilutions with concentration on the x axis (log scale) versus absorbance on the Y axis (linear). The concentration of the sample is interpolated from this standard curve. Nonlinear regression is often used to fit standard curves generated by ELISA assay which is based on competitive binding. The assaying compound competes for binding to an enzyme or antibody with a labeled compound. Therefore the standard curve is described by equations for competitive binding. 191 Chapter 14 – Appendices One-site competitive binding curve is tried first. If that doesn’t fit the data well, sigmoid curves are tried, by entering the X values as the logarithms of concentrations. Choice of an equation is important when using nonlinear regression, if the equation does not describe a model that makes scientific sense; the results of nonlinear regression won’t make sense either. With standard curve calculations, the choice of an equation is less important because all one has to do is assess visually that the curve nicely fits the standard points. 192 Chapter 14 – Appendices Appendix 2. Human Amylin (Total) ELISA KIT and Assay Procedure The Human Amylin ELISA is a monoclonal antibody-based sandwich immunoassay for determining total amylin levels in human plasma. The capture antibody recognizes reduced human amylin and human amylin acid (deamidated amylin) but not the 1-20 fragment of amylin. The detection antibody binds to reduced or unreduced human amylin but not amylin acid and is complexed with streptavidin-alkaline phosphatase to form a sandwich. The substrate, 4-Methylumbelliferyl Phosphate (MUP), is applied to the completed sandwich and the fluorescent signal, monitored at 355 nm/460 nm, is proportional to the amount of amylin present in the sample. After receipt of the kit, all components of the kit were stored at 2-8 degrees centigrade. The assay is run in duplicate using 50 µl of assay buffer and 50 µl of Standard, Control, or Sample in each well. The assay procedure is as follows: The concentrated TBS (Tris Buffered Saline) wash buffer is diluted 10 fold by mixing the entire contents of the 10 X wash buffer with 270 ml deionized water. Standards and QCI and QC2 (lyophilized) vials is reconstituted with 250 µl of deionoized water. The solution is mixed gently for at least 5 minutes to assure complete reconstitution. The microtitre assay plate is removed from the foil pouch and each well is filled with 300 µl of diluted TBS wash buffer which is incubated at room temperature for 10 minutes without shaking. Wash buffer is decanted from the plate and the residual amount removed from all wells by inverting the plate and tapping it smartly onto absorbent towels several times. The well is not allowed to dry before proceeding to the next step. 50 µl of assay buffer is added to each well. 50 µl Standards, Samples and Controls are added in duplicates. The wells are orientated as highlighted below. The plate is sealed and incubated at room temperature on the shaker for one hour. It is important to note that the incubation start time is the time as plate was loaded on the shaker, not from the time one started loading the plate with samples. The residual amount from all wells is decanted and removed by 193 Chapter 14 – Appendices inverting the plate and tapping it smartly onto absorbent towels several times. Microtitre plate arrangements of wells is as shown below 1 2 3 4 5 6 7 8 9 10 11 12 A 0 pM 0 pM 1 pM 1 pM 2 pM 2 pM 5 pM 5 pM 10 pM 10 pM 40 pM 40 pM B 100 pM 100 pM QC1 QC1 QC2 QC2 Sam ple 1 Sam ple 1 Samp le 2 Sam ple 2 Sam ple 3 Sa mpl e3 C D E F G H The plate is washed 3 times with 300 µl per well TBS wash buffer. The plate is decanted and tapped after each wash to remove residual buffer. 100 µl detection antibody is added to each well, plate is covered with sealer, and incubated on the shaker at room temperature for 2 hours. Near the completion of this incubation step, the substrate is hydrated by adding 1 ml deionized water to 10 mg, mixed well, and allowed to stand for 15 minutes with occasional mixing to assure complete dissolution. 105 µl is removed from the reconstituted substrate and added to the 21 ml vial of substrate diluent, mixed well, referred to as substrate solution from here on. Detection antibody is decanted and the residual amount removed from all wells by inverting the plate and tapping it smartly onto absorbent towels several times. 194 Chapter 14 – Appendices The plate is washed 3 times with 300 µl per well TBS wash buffer, decanted and tapped after each wash, to remove residual buffer. 100 µl substrate solution is added to each well and incubated for 15 minutes at room temperature in the dark without shaking. A reading is taken and the RFU (Relative Fluorescence Unit) of the top standard point is noted; when the reading is 2000 RFU or greater, 50 µl stop solution is added, gently mixed, and read on the fluorescence plate reader after 5 minutes. The RFU can be fitted directly to the concentration. If the curve fitting software is available, the best fit can be obtained with a linear-linear spline fit. Since this assay is a direct ELISA, the RFU is directly proportional to the concentration of total human amylin in the sample. It is important to note that when sample volumes assayed differ from 50 µl, an appropriate mathematical adjustment must be made to accommodate for the dilution factor (e.g. if 25 µl of sample is used, then calculated data must be multiplied by 2). 195 Chapter 14 – Appendices Appendix 3. Characteristics of Human Amylin (Total) ELISA Kit The human amylin ELISA kit had the following assay characteristics: Sensitivity: The lowest level of total human amylin that could be detected by this assay is 1 pM (50 µl plasma sample size). Performance: The parameters expressed as mean ± standard deviation of assay performance are: Parameters Mean ± SD ED 80 82 ± 2 pM ED 50 54 ± 5 pM ED 20 2 6± 4 pM Crossreactivity: The cross reactivity of the assay is as follows: Peptides Percentage Human Glucagon <1% Human GLP-1 <1% Human Insulin <1% Human Pancreatic Polypeptide <1% Human Adrenomedullin 1% Human Calcitonin <1% Calcitonin Gene Related Peptide <1% Precision: The assay variation of Linco Human Amylin ELISA kits were studied at three different spiked concentrations of amylin in three different human plasma samples. The within variations was the mean from four duplicate determinations in a single assay. The between variation was the mean value of the mean of four duplicate determinations in each plasma across four assays. Within and between assay variation is as follows: 196 Chapter 14 – Appendices Sample No 1 Amylin Added pM 20 50 80 Within % CV 2.6 5.0 14.8 Between % CV 11.9 12.9 11.5 2 20 50 80 11.5 2.5 7.9 17.4 17.8 16.2 3 20 50 80 7.8 5.4 7.3 17.6 11.9 13.0 Recovery: Varying concentrations of Human Amylin were added to three human plasma samples and the amylin content was determined in four different ELISA assays. The % of recovery=observed amylin concentration/expected amylin concentration x 100%. Spike and recovery of total human amylin in human plasma was as follows: Sample 1 Sample Concentration (pM) 3.25 Amylin % of Recovery 20 50 80 104 99 91 2 2.56 20 50 80 96 89 89 3 11.23 20 50 80 101 87 90 Linearity: Three human plasma samples with the indicated sample volumes were assayed in four different assays. Required amount of assay buffer was added to compensate for lost volumes below 50 µl. The resulting dilution factors of 1.0, 1.25, 2.0, and 5.0 representing 50 µl, 40 µl, 25 µl, and 10 µl sample volumes assayed, respectively, were applied. The effect of plasma dilution was as follows: 197 Chapter 14 – Appendices Sample No 1 Volume Sampled 50 40 25 10 Expected pM Observed pM 22.32 22.32 21.13 19.98 18.55 100 95 90 83 2 50 40 25 10 25.34 25.35 22.30 20.54 21.05 100 88 81 83 3 50 40 25 10 20.54 20.54 20.85 15.96 19.65 100 101 78 96 198 %of Expected Chapter 14 – Appendices Appendix 4. Research protocol for ethics approval, South Sheffield Research Ethics Committee Title: Amylin peptide: Association with feed intolerance in preterm infants and IMDM- Observational & Case-Control Study Study Sponsor: University Hospitals of Sheffield NHS Trust Study Summary Title Amylin peptide: Association with feed intolerance in preterm infants & IMDM Short Title Amylin peptide Methodology Prospective Observational Study and CaseControl Study Study Duration 12 months Study Centres Single Centre (Study A & B ) & Two Centre (Study C) Objectives To determine serum amylin levels in well preterm and term infants (Study A), infants of diabetic mothers (Study B) and preterm infants with feed intolerance (Study C). Number of Samples/Participants Study A=200 samples Study B= 21 IDM Study C= 20 nTOL and 20 TOL Main Inclusion Criteria 1. Infants ≥ 24 weeks gestation 2. Infants of diabetic mothers 3. Preterm infants (24-36 weeks) admitted to the neonatal intensive care unit and requiring gastric tube feeding Statistical Methodology Median and interquartile ranges and nonparametric tests. Correlation Coefficient will be performed to evaluate the relationship between variables. Bland altman test will be applied for method comparison between umbilical arterial and umbilical venous blood sampling method. Outcome variable Serum amylin levels in pmol/l 199 Chapter 14 – Appendices Background: The goal of neonatal nutrition is the attainment of normal growth and development. However impairment of gastric motility is common in critically ill patients (1) and failure to establish enteral feeds in neonates necessitates the use of parenteral nutrition which carries a significant increased risk of morbidity and mortality (2). There is scant information on the mechanisms relating to feed intolerance in these patients, therefore limiting the therapeutic options. Insight into the regulation of gastric motility has been advanced by the recent discovery of ‘amylin’, a novel 37 amino-acid peptide hormone from the calcitonin gene-related peptide family. As a potent inhibitor of gastric emptying it plays an important physiological role in the control of carbohydrate absorption by regulating the efflux from the stomach to the small bowel. The pathophysiological role of amylin is well documented in adult patients with diabetes mellitus (1), where inappropriately high levels have been reported with delayed gastric emptying. There is no published data on amylin levels and its role in neonatal population. We aim to determine normal amylin levels in neonatal population and investigate its role in infants born to mothers whose pregnancies were complicated by diabetes (IMDM) & preterm infants with feed intolerance. Rationale and significance of the study: Up to 67% of infants weighing less than 1500 g (1-3) experience feed intolerance resulting in prolonged parenteral fluid therapy, increased hospital stay, increased risk of sepsis and consequently increased cost of treatment (4). Feed intolerance is also a major problem in IMDM (5). An increased prefeed gastric residual volume is a commonly used measure of feed intolerance in newborn infants. Decreased gastrointestinal motility can be multifactorial in origin, we hypothesize that amylin may have an important role to play in its pathophysiology. Amylin is novel “brain-gut peptide” which is co-secreted with insulin from the pancreas (6). In conjunction to playing a role in glucose homeostasis, it is a potent inhibitor of gastric emptying (7). A study in critically ill infants and children showed elevated amylin levels which correlated with delayed gastric emptying and feed intolerance (8). 200 Chapter 14 – Appendices We aimed to investigate the role of amylin in newborn infants with feed intolerance which could potentially form the basis of a subsequent clinical trial. Research question: Is amylin elevated in neonates with feed intolerance (delayed gastric emptying)? Aims and Objectives: 1. To determine reference range of amylin at birth (umbilical cord) and at 5-7 days (Guthrie) in term and preterm neonates (Study A). 2. To determine amylin levels in IMDM (Study B). 3. To determine amylin in preterm neonates with feed intolerance (study C). Study design: Prospective observational and case-control study to be conducted simultaneously as 3 sub-studies. Study A- To establish normal ranges of cord and postnatal amylin levels, mothers will be recruited antenatally for umbilical cord blood and inpatient Guthrie blood. Study B- All infants of diabetic mothers will be recruited for cord bloods, Guthrie bloods, and opportunistic bloods taken at the time of routine glucose measurement. Study C- Preterm infants admitted to neonatal intensive care unit and requiring tube feeding will be recruited. Amylin levels of feed intolerant infants (case) will be compared to feed tolerant infants (control) Study population: Study A-Term and preterm infants > 24 weeks of gestation, Study B-Term IMDM and preterm IMDM > 24 weeks of gestation and Study CPreterm infants > 24 weeks of gestation Exclusion Criteria 1. Infants born less than 24 weeks gestation 2. Congenital malformation 3. Chromosomal anomaly 4. Structural gastrointestinal abnormalities and necrotising enterocolitis 5. Haemoglobin less than 7 g/dl 6. Breast feeding where feed volume cannot be determined Primary outcome measure: Serum amylin levels (pmol/l) 201 Chapter 14 – Appendices Definition of feed tolerance: Feed intolerance will be defined as prefeed gastric residual volumes of greater than 50% of the previous feed on 2 consecutive occasions (4). Method of collection and analysis: 0.5 ml-1ml blood will be drawn into a tube containing EDTA with 500 IU of aprotinin per millilitre of blood and kept on ice before separation and stored at -70˚C. All samples will be analysed for total amylin levels using monoclonal antibody-based sandwich immunoassay. The amylin ELISA kit is validated for the measurement of total amylin from EDTA human plasma samples. It has sensitivity to 1 pM and a standard range of 1100 pM. Were possible, plasma immunoreactive insulin will be measured by ELISA. Blood glucose will be measured by the glucose oxidase method. The assays will be performed by technical team blinded to the patient data led by Mr R Fraser. Setting: Labour ward and neonatal intensive care unit, Jessop wing, Royal Hallamshire Hospital. Study C will also be conducted at the neonatal intensive care unit, Doncaster General Hospital. Consent: Parents will be recruited prior to delivery and written consent obtained. Study Plan: Estimated sample volume is 0.5-1.0 ml. Prefeed gastric aspirates are recorded as part of routine neonatal practise. Data will be collected with reference to method of nutrition, demographic data and medication at the time of sample collection. Study A: Mothers of babies having normal, preterm and LSCS deliveries will be recruited. Cord blood will be obtained immediately after delivery of these babies. A repeat sample will be taken at the same time as routine Guthrie from babies who are inpatients. These will be analysed for amylin and insulin. Study B: Cord and Guthrie blood and a daily sample taken at the time routine blood sugar (hypoglycaemia protocol) of infants of diabetic mothers, and will be analysed for amylin and insulin. Study C: Preterm infants with apparent feed intolerance will be recruited when delayed gastric emptying is observed. Samples for amylin, insulin and glucose will be taken at the next routine phlebotomy. 202 Chapter 14 – Appendices Analysis: A sample size of 200 in study A and 20 each in study B and study C will have a 90% power to detect a difference in means of 15 (the difference between a group 1 mean of 37.7 and group 2 mean of 22.7) assuming that the common standard deviation is 13 using a two group t-test with a 0.05 two sided significance level. Benefits and risks: There will be no benefit to the babies included in the trial. However there are no known risks involved. The information we get from this study may help us to understand the mechanisms of delayed emptying of the stomach. Confidentiality: Patient data will be allocated a unique number and only the research team will have access to family identities. All patient information will be stored in locked filing cabinet and patient information will be stored on password protected PC’s at Jessop wing, Royal Hallamshire hospital. Resource Requirements Amylin Kits £270/Kit £1080 Serum tubes £50 Technical staff time £0 Trasylol £10 Dissemination and Outcome: The data will be presented at local, national and international meetings. The results will be published in pediatrics research journals. References 1. Dive A et al. Crit Care Med 1994;22:441-447 2. Yu VY. Acta Med Port 1997;10(2-3):185-196 3. Premji SS et al. Cisapride: A review of the evidence supporting its use in premature infants with feeding intolerance. Neonatal Netw 1997; 16: 1721 4. Gross SJ. Growth and biochemical response of preterm infants fed human milk or modified infant formula. N Engl J Med 1983; 308:237-241. 5. Gross SJ et al. Feeding the low birth weight infant. Clin Perinatol 1993; 20: 193-209. 203 Chapter 14 – Appendices 6. Reddy PS et al. Double blind placebo controlled study on prophylactic use of cisapride on feed intolerance and gastric emptying in preterm neonates. Indian Pediatr 2000; 37: 837-844 7. Kitzmiller J L. Am J Obstet Gynecol 1978; 131: 560. 8. Ludvig B et al. Amylin: History and overview. Diabetic Med 1997; 14: S9S13 9. Macdonald IA. Amylin and the gastrointestinal tract. Diabetic Med 1997; 14: S24-S28. 10. Mayer AP. Amylin is associated with delayed gastric emptying in critically ill children. Intensive Care Medicine. 2002; 28(3): 336-340. 204 Chapter 14 – Appendices Appendix 5. Research protocol for ethics approval, Doncaster Research Ethics Committee Amylin peptide: Association with feed intolerance in preterm infants- A CaseControl Study Study Sponsor: Doncaster General Hospital NHS Trust Study Summary Title Amylin peptide: Association with feed intolerance in preterm infants Short Title Amylin peptide Methodology Case-Control Study Study Duration 12 months Study Centre Single Centre Objectives To determine serum amylin levels in preterm infants with feed intolerance. Number of Samples/Participants 20 nTOL and 20 TOL Main Inclusion Criteria Preterm infants (24-36 weeks) admitted to the neonatal intensive care unit and requiring gastric tube feeding Statistical Methodology Median and interquartile ranges and nonparametric tests. Corelation Coefficient will be performed to evaluate the relationship between variables. Outcome variable Serum amylin levels in pmol/l Background: The goal of neonatal nutrition is the attainment of normal growth and development. However impairment of gastric motility is common in critically ill patients (1). Failure to establish enteral feeds in neonates necessitates the use of parenteral nutrition which carries significantly increased risk of morbidity and mortality (2). There is scant information on the mechanisms relating to feed intolerance in these patients, therefore limiting the therapeutic options. 205 Chapter 14 – Appendices Insight into the regulation of gastric motility has been advanced by the discovery of ‘amylin’, a novel 37 amino-acid peptide hormone from the calcitonin generelated peptide family. As a potent inhibitor of gastric emptying it plays an important physiological role in the control of carbohydrate absorption by regulating the efflux from the stomach to the small bowel. The pathophysiological role of amylin is well documented in adult patients with diabetes mellitus (1), where inappropriately high levels have been reported with delayed gastric emptying. We have previously established normal serum amylin levels in umbilical cord and Guthrie blood samples from healthy preterm and term new-born infants. This study is planned to investigate the role of amylin in preterm infants with feed intolerance. Rationale and significance of the study: Feed intolerance has been reported in up to 67% of infants weighing less than 1500 g (1-3) resulting in prolonged parenteral fluid therapy, increased hospital stay, increased risk of sepsis and consequently increased cost of treatment (4). Decreased gastrointestinal motility can be multifactorial in origin, we hypothesize that amylin may have an important role to play in its pathophysiology. Amylin is a novel “brain-gut peptide” which is co-secreted with insulin from the pancreas (6). In conjunction to playing a role in glucose homeostasis, it is a potent inhibitor of gastric emptying (7). A study examining critically ill infants and children with feed intolerance showed elevated serum amylin levels (8). We aim to establish the association of feed intolerance to elevated amylin levels in the neonatal population which could potentially form the basis of a subsequent clinical trial using a commercially available amylin antagonist. Research question: Is amylin elevated in neonates with delayed gastric emptying? Aim and Objectives: To establish amylin levels in preterm neonates with delayed gastric emptying (feed intolerance). 206 Chapter 14 – Appendices Study design: This is a case-control study. The cases will be preterm infants admitted to neonatal intensive care unit who subsequently develop feed intolerance. Preterm infants without feed intolerance matched for birth weight, gestation and postnatal age will be controls. Study population: Preterm infants between 24-36 weeks of gestation Exclusion Criteria 1. Congenital malformation 2. Chromosomal anomaly 3. Structural gastrointestinal abnormalities and necrotising enterocolitis 4. Breast feeding where feed volume cannot be determined 5. Infant born to mother whose pregnancy is complicated by diabetes 6. Primary outcome: Serum amylin levels in infants with and without feed intolerance. Definition of feed intolerance: In this population feed intolerance will be defined as, prefeed gastric residual volume (GRV) of greater than 50% of the previous feed (4), increase in the abdominal girth from baseline by more than 2 cm in the region of the epigastrium over 24 hours by regular monitoring and vomiting. Method of collection and analysis: Blood for amylin and insulin will be drawn into a tube containing EDTA with 500 IU of aprotinin per millilitre of blood and kept on ice before separation and stored at -70˚C. The total amylin levels in human plasma are assayed using an ELISA monoclonal antibody-based sandwich immunoassay. The amylin ELISA kit is validated for the measurement of total amylin from EDTA human plasma samples. It has sensitivity to 1 pM and a standard range of 1-100 pM. Plasma immunoreactive insulin will be measured by ELISA. Blood glucose will be measured by the glucose oxidase method. The assays will be done by technical team under R Fraser who will be blinded to the patient data. Setting: Neonatal intensive care unit, Doncaster Royal Infirmary 207 Chapter 14 – Appendices Consent: Parents of infants admitted to the neonatal unit and requiring tube feeding will be approached for consent. Study Plan: All the infants admitted to the neonatal intensive care unit and requiring tube feeding will be eligible for the study. The feeding regime of the neonates will be dictated by the local guideline. Expressed breast milk (EBM) is encouraged, and formula feed is used if breast milk is not available. The neonates are fed every hour and the volumes increased as clinically tolerated but not more than 20mls/kg birth weight/day. Every four hours pre-feed GRV will be recorded as a percentage of the previous feed volume. Abdominal girth, colour of gastric residuals and emesis will also be noted. If this GRV is greater than 50% on two consecutive occasions, neonates will be classified as feedintolerant (nTOL). The further management of these neonates is up to the attending neonatologist. Researcher or responsible consultant will identify preterm infants introduced to gastric tube feeding who are receiving > 30 ml/kg or > 25% of total fluid intake as enteral feeds. Parents will be approached for consent and information leaflets provided. Blood for amylin will be collected within 2 hours of the second big (> 50%) prefeed GRV in nTOL neonates. 0.5 ml blood will be collected from capillary, vein, or indwelling arterial catheters as deemed necessay for routine collection. Feed-tolerant (TOL) neonates matched for gestational age, birth weight and postnatal age will serve as controls. 0.5 ml blood for amylin in these neonates will be collected during their next routine collection. Data will be collected with reference to method of nutrition, demographic data and medication at the time of sample collection. Analysis: A sample size of 20 each in nTOL and TOL will have a 90% power to detect a difference in means of 15 (the difference between a group 1 mean of 37.7 and group 2 mean of 22.7) assuming that the common standard deviation is 13 using a two group t-test with a 0.05 two sided significance level. Benefits and risks: There will be no benefit to the babies included in the trial. However there are no known risks involved. The information we get from this study may help us to understand the mechanisms of delayed emptying of the stomach. 208 Chapter 14 – Appendices Confidentiality: Patient data will be allocated a unique number and only the research team will have access to family identities. All patient information will be stored in locked filing cabinet and patient information will be stored on password protected PC’s at Women’s Hospital, Doncaster Royal Infirmary Resource Requirements Amylin Kits £270/Kit £270 Serum tubes £50 Trasylol £10 Dissemination and Outcome: The data will be presented at local, national and international meetings. The results will be published in paediatrics research journals. References 1. Dive A et al. Crit Care Med 1994;22:441-447 2. Yu VY. Acta Med Port 1997;10(2-3):185-196 3. Premji SS et al. Cisapride: A review of the evidence supporting its use in premature infants with feeding intolerance. Neonatal Netw 1997; 16: 1721 4. Gross SJ. Growth and biochemical response of preterm infants fed human milk or modified infant formula. N Engl J Med 1983; 308:237-241. 5. Gross SJ et al. Feeding the low birth weight infant. Clin Perinatol 1993; 20: 193-209. 6. Reddy PS et al. Double blind placebo controlled study on prophylactic use of cisapride on feed intolerance and gastric emptying in preterm neonates. Indian Pediatr 2000; 37: 837-844 7. Kitzmiller J L. Am J Obstet Gynecol 1978; 131: 560. 8. Ludvig B et al. Amylin: History and overview. Diabetic Med 1997; 14: S9S13 9. Macdonald IA. Amylin and the gastrointestinal tract. Diabetic Med 1997; 14: S24-S28. 10. Mayer AP. Amylin is associated with delayed gastric emptying in critically ill children. Intensive Care Medicine. 2002; 28(3): 336-340. 209 Chapter 14 – Appendices Appendix 6. Child participation information leaflet (Study A & B) CHILD PARTICIPATION INFORMATION LEAFLET Neonatal Intensive Care Unit, Jessop Wing, Royal Hallamshire Hospital Study title: Amylin peptide: Association with feed intolerance in preterm infants and IMDM. What is this study about? You and your baby are being invited to take part in a research study. Before you decide whether to take part, it is important for you to understand why the research is being done and what it will involve. Please take time to read the following information carefully and discuss it with others if you wish. Ask us if there is anything that is not clear or you would like more information. Please take time to decide whether or not you wish to take part. What is the purpose of the study? Many critically ill babies admitted to the intensive care unit have major problems tolerating normal milk feeds. This is especially common in those babies born early (prematurely) and babies born to mothers with sugar intolerance (diabetes mellitus). We are presently investigating possible reasons why this may occur. One explanation is that a substance called AMYLIN present in the blood stream may be responsible. Amylin is a newly discovered hormone that potently inhibits the function of the stomach. Because good nutrition is crucial to the healing process in sick babies we are conducting a study to measure Amylin levels in the hope that this may shed some light on this difficult problem. If a relationship can be shown between blood Amylin levels and poor stomach function we could potentially develop treatments in the future to promote feeding, improve nutrition and hopefully hasten recovery time in critically ill babies. To study this chemical we aim to measure it in all babies born at this hospital at the time of routine blood taking for neonatal screening. Why has my baby been chosen? 210 Chapter 14 – Appendices Your baby has been chosen because if he/she is not tolerating feeds or is slow to feed, we are trying to find out if the feed intolerance is due to increased blood amylin level. Do I have to take part? It is up to you to decide whether or not to take part. If you do decide to take part you will be given this information sheet to keep and be asked to sign a consent form. If you decide to take part you are still free to withdraw at any time and without giving a reason. This will not affect the standard of care your baby receives. What will happen if my baby takes part? If you decide to take part in the study the only change from routine management is that 0.5 ml (about 8 drops) of your baby’s blood would be taken during routine blood sampling, so causing no added distress or pain. Additionally a sample would be taken from umbilical cord after delivery (this causes no added distress or discomfort to your baby as the sample is taken after the cord is cut). What are the possible disadvantages and risks of taking part? We will collect no more than 0.5 ml blood. This is safe amount to take from your baby. What are the possible benefits of taking part? There will be no direct health benefit to your baby. What if something goes wrong? If you have any cause to complain about any aspect of the way in which you have been approached or treated during the course of this study, the normal National Health Service complaints mechanisms are available to you and are not compromised in any way because you have taken part in a research study. If you have any complaints or concerns please contact the project co-ordinator: (Dr Coombs on telephone 01142268387). Otherwise you can use the normal 211 Chapter 14 – Appendices hospital complaints procedure and contact the following person: (Chris Welsh, Medical Director STH, 8 Beech Hill Road, Sheffield, S10 2SB, 0114 271 2178). Will my baby’s taking part in this study be kept confidential? All information, which is collected, about your baby during the course of the research will be kept strictly confidential. Any information about your baby, which leaves the hospital/surgery, will have your name and address removed so that you cannot be recognised from it. What will happen to the results of the research study? When the study finishes all the results will be analysed, which may lead to a publication in a medical journal. At no time will any identifying information will be published and all records will remain confidential. If you wish we will forward you the published report of the study. Who is organising and funding the research? The research is being organised by the neonatal intensive care unit at Jessops Wing, Royal Hallamshire hospital. There is no outside funding or financial incentive to this study. Who has reviewed the study? The study has been reviewed and approved by South Sheffield Ethics Committee. Who can I contact if I want to know more about the study? Drs V Kairamkonda & A Deorukhkar, Neonatal intensive care unit, Jessop Wing, Royal Hallamshire Hospital, Sheffield S10 2SF, Tel: 01142268322, Fax: 01142268449. Thank you for reading this information sheet. 212 Chapter 14 – Appendices Appendix 7. Child participation information leaflet (Study C) Royal Hallamshire Hospital CHILD PARTICIPATION INFORMATION LEAFLET Neonatal Intensive Care Unit, Jessop Wing, Royal Hallamshire Hospital Study title: Amylin peptide: Association with feed intolerance in preterm infants. What is the purpose of the study? Many critically ill babies admitted to the intensive care unit have major problems tolerating normal milk feeds. This is especially common in those babies born early (prematurely) and babies born to mothers with sugar intolerance (diabetes mellitus). We are presently investigating possible reasons why this may occur. One explanation is that a substance called AMYLIN present in the blood stream may be responsible. Amylin is a newly discovered hormone that potently inhibits the function of the stomach. Because good nutrition is crucial to the healing process in sick babies we are conducting a study to measure Amylin levels in the hope that this may shed some light on this difficult problem. If a relationship can be shown between blood Amylin levels and poor stomach function we could potentially in the future use drugs to block the action of Amylin in order to promote feeding, improve nutrition and hopefully hasten recovery time in critically ill babies. To study this chemical we aim to measure it in all preterm babies admitted to the intensive care with feed intolerance or who subsequently develop feed intolerance. Why has my baby been chosen? Your baby has been chosen because if he/she is not tolerating feeds or is slow to feed, we are trying to find out if the feed intolerance is due to increased blood amylin level. Do I have to take part? It is up to you to decide whether or not to take part. If you do decide to take part you will be given this information sheet to keep and be asked to sign a consent form. If you decide to take part you are still free to withdraw at any time and 213 Chapter 14 – Appendices without giving a reason. This will not affect the standard of care your baby receives. What will happen if my baby takes part? If you decide to take part in the study the only change from routine management is that 0.5ml (about 8 drops) of your baby’s blood would be taken during routine blood sampling, so causing no added distress or pain. What are the side effects of taking part? None What are the possible disadvantages and risks of taking part? None What are the possible benefits of taking part? None What if something goes wrong? If you have any cause to complain about any aspect of the way in which you have been approached or treated during the course of this study, the normal National Health Service complaints mechanisms are available to you and are not compromised in any way because you have taken part in a research study. If you have any complaints or concerns please contact the project co-ordinator: (Dr Coombs on telephone 01142268387). Otherwise you can use the normal hospital complaints procedure and contact the following person: (Chris Welsh, Medical Director STH, 8 Beech Hill Road, Sheffield, S10 2SB, 0114 271 2178). Will my baby’s taking part in this study be kept confidential? All information, which is collected, about your baby during the course of the research will be kept strictly confidential. Any information about your baby, which leaves the hospital/surgery, will have your name and address removed so that you cannot be recognised from it. 214 Chapter 14 – Appendices What will happen to the results of the research study? When the study finishes all the results will be analysed, which may lead to a publication in a medical journal. At no time will any identifying information will be published and all records will remain confidential. If you wish we will forward you the published report of the study. Who is organising and funding the research? The research is being organised by the neonatal intensive care unit at Jessops Wing, Royal Hallamshire Hospital. There is no outside funding or financial incentive to this study. Who has reviewed the study? The study has been reviewed and approved by South Sheffield Ethics Committee. Contacts for Further Information Drs V Kairamkonda & A Deorukhkar, Neonatal intensive care unit, Jessop Wing, Royal Hallamshire Hospital, Sheffield S10 2SF, Tel: 01142268322, Fax: 01142268449 Thank you for reading this information sheet. 215 Chapter 14 – Appendices Appendix 8. Child participation information leaflet (Study C) Doncaster Royal Infirmary CHILD PARTICIPATION INFORMATION LEAFLET Neonatal Intensive Care Unit, Doncaster Royal Infirmary, Doncaster Study title: Amylin peptide: Association with feed intolerance in preterm infants. What is the purpose of the study? Many critically ill babies admitted to the intensive care unit have major problems tolerating normal milk feeds. This is especially common in those babies born early (prematurely) and babies born to mothers with sugar intolerance (diabetes mellitus). We are presently investigating possible reasons why this may occur. One explanation is that a substance called AMYLIN present in the blood stream may be responsible. Amylin is a newly discovered hormone that potently inhibits the function of the stomach. Because good nutrition is crucial to the healing process in sick babies we are conducting a study to measure Amylin levels in the hope that this may shed some light on this difficult problem. If a relationship can be shown between blood Amylin levels and poor stomach function we could potentially in the future use drugs to block the action of Amylin in order to promote feeding, improve nutrition and hopefully hasten recovery time in critically ill babies. To study this chemical we aim to measure it in all preterm babies admitted to the intensive care with feed intolerance or who subsequently develop feed intolerance. Why has my baby been chosen? Your baby has been chosen because if he/she is not tolerating feeds or is slow to feed, we are trying to find out if the feed intolerance is due to increased blood amylin level. Do I have to take part? It is up to you to decide whether or not to take part. If you do decide to take part you will be given this information sheet to keep and be asked to sign a consent form. If you decide to take part you are still free to withdraw at any time and 216 Chapter 14 – Appendices without giving a reason. This will not affect the standard of care your baby receives. What will happen if my baby takes part? If you decide to take part in the study the only change from routine management is that 0.5ml (about 8 drops) of your baby’s blood would be taken during routine blood sampling, so causing no added distress or pain. What are the side effects of taking part? None What are the possible disadvantages and risks of taking part? None What are the possible benefits of taking part? None What if something goes wrong? If you have any cause to complain about any aspect of the way in which you have been approached or treated during the course of this study, the normal National Health Service complaints mechanisms are available to you and are not compromised in any way because you have taken part in a research study. If you have any complaints or concerns please contact the project co-ordinator: (Dr Anjum Deorukhkar on telephone 01302 366666 ext 3166). Will my baby’s taking part in this study be kept confidential? All information, which is collected, about your baby during the course of the research will be kept strictly confidential. Any information about your baby, which leaves the hospital/surgery, will have your name and address removed so that you cannot be recognised from it. What will happen to the results of the research study? When the study finishes all the results will be analysed, which may lead to a publication in a medical journal. At no time will any identifying information will be 217 Chapter 14 – Appendices published and all records will remain confidential. If you wish we will forward you the published report of the study. Who is organising and funding the research? The research is being organised by the neonatal intensive care unit at Women’s Hospital Doncaster Royal Infirmary, Doncaster. There is no outside funding or financial incentive to this study. Who has reviewed the study? The South Sheffield Ethics Committee and the Doncaster Research Ethics Committee. Contacts for Further Information Dr. A Deorukhkar, Neonatal intensive care unit, Women’s Hospital DRI Doncaster, Telephone: 01302 366666 ext 3166 Thank you for reading this information sheet. 218 Chapter 14 – Appendices Appendix 9. Study A & B Consent form CONSENT FORM FOR PARTICIPATION OF CHILD Title of Project: Amylin peptide: Association with feed intolerance in preterm infants and IMDM. Patient Identification Number: Centre Number: Study Number: Name of Researchers: V Kairamkonda, A Mayer, A Deorukhkar, R Coombs, R Fraser Please initial box I confirm that I have read and understand the information sheet Version 14/8/00 for the above study and have had the opportunity to ask questions. I understand that my participation is voluntary and that I am free to withdraw at any time, without giving any reason, without my medical care or legal rights being affected. I understand that sections of any of my medical notes may be looked at by responsible individuals from research team or from regulatory authorities where it is relevant to my taking part in research. I give permission for these individuals to have access to my records. I agree to take part in the above study. ________________________ Name of Patient _______________ ___________________ Date Signature _________________________ Name of Person taking consent ________________ ___________________ Date Signature Original to be kept in baby’s medical notes; 1 copy to parent; 1 copy for research file 219 Chapter 14 – Appendices Appendix 10. Study C Consent form CONSENT FORM FOR PARTICIPATION OF CHILD Title of Project: Amylin peptide: Association with feed intolerance in preterm infants. Patient Identification Number: Centre Number: Study Number: Name of Researchers: V Kairamkonda, A Mayer, A Deorukhkar, R Coombs, R Fraser Please initial box I confirm that I have read and understand the information sheet Version 14/8/00 for the above study and have had the opportunity to ask questions. I understand that my participation is voluntary and that I am free to withdraw at any time, without giving any reason, without my medical care or legal rights being affected. I understand that sections of any of my medical notes may be looked at by responsible individuals from research team or from regulatory authorities where it is relevant to my taking part in research. I give permission for these individuals to have access to my records. I agree to take part in the above study. ________________________ Name of Patient _______________ ___________________ Date Signature _________________________ Name of Person taking consent ________________ ___________________ Date Signature Original to be kept in baby’s medical notes; 1 copy to parent; 1 copy for research file 220 Chapter 14 – Appendices Appendix 11. Advert for Study A AMYLIN STUDY Dear Parents Feed intolerance is a common problem in babies admitted to neonatal intensive care unit. Amylin is a newly discovered hormone that potently inhibits the function of the stomach. We are conducting a study to measure amylin levels in the hope that this may shed some light on this difficult problem. The study involves taking blood from your baby at birth from discarded umbilical cord and at day 5 routine Guthrie (few drops) If you would like further information or interested in taking part, please contact staff on the unit OR Dr Venkatesh Kairamkonda, Specialist Registrar 0114228456/68356 OR Dr Anjum Deorukhkar, clinical research fellow 0114228456/68356. 221 Chapter 14 – Appendices Appendix 12. Advert for Study B AMYLIN STUDY Dear Parents Feed intolerance is a common problem in babies born to mothers whose pregnancy was complicated by diabetes. Amylin is a newly discovered hormone that potently inhibits the function of the stomach. We are conducting a study to measure amylin levels in the hope that this may shed some light on this difficult problem. The study involves taking blood from your baby at birth from discarded umbilical cord and at day 5 routine Guthrie (few drops) If you would like further information or interested in taking part, please contact staff on the unit OR Dr Venkatesh Kairamkonda, Specialist Registrar 0114228456/68356 OR Dr Anjum Deorukhkar, clinical research fellow 0114228456/68356. 222 Chapter 14 – Appendices Appendix 13. Advert for Study C AMYLIN STUDY Dear Parents Feed intolerance is a common problem in babies admitted to neonatal intensive care unit. Amylin is a newly discovered hormone that potently inhibits the function of the stomach. We are conducting a study to measure amylin levels in the hope that this may shed some light on this difficult problem. The study involves taking a small sample of your baby’s blood during routine collection (few drops). If you would like further information or interested in taking part, please contact staff on the unit OR Dr Venkatesh Kairamkonda, Specialist Registrar 0114228456/68356 OR Dr Anjum Deorukhkar, clinical research fellow 0114228456/68356. 223 Chapter 14 – Appendices Appendix 14. Case report form Study A CASE REPORT FORM STUDY A Study ID Date Baby details Patient Sticker Name Male/Female Date of Birth Hospital Number Address Baby Details Birth weight Gestational age Growth Type AGA SGA LGA Cord blood sample Arterial Venous Both Postnatal blood sample yes no Guthrie sample yes no APGAR APGAR1 APGAR5 Resuscitation yes no ETVentilation/CPAP yes no Sepsis yes no PROM yes no Chorioamnionitis yes no Antenatal steroids yes no Date of blood sampling Number of blood samples Mode of delivery Maternal Details Maternal Diagnosis Medications 224 APGAR10 Chapter 14 – Appendices Appendix 15. Case report form Study B CASE REPORT FORM STUDY B Study ID Date Baby details Patient Sticker Name Male/Female Date of Birth Hospital Number Address Baby Details Birth weight Gestational age Growth Type AGA SGA LGA Cord blood sample Arterial Venous Both Postnatal blood sample yes no Guthrie sample yes no Mode of delivery CS Instrumental Normal APGAR APGAR1 APGAR5 APGAR10 Resuscitation yes no ETVentilation/CPAP yes yes Sepsis yes no Feed intolerance yes no Diabetes Type GDM IDDM PROM yes no Chorioamnionitis yes no Antenatal steroids yes no Date of blood sampling Number of blood samples Maternal Details Maternal Diagnosis Medications 225 NIDDM Chapter 14 – Appendices Appendix 16. Case report form Study C CASE REPORT FORM STUDY C Study ID Date Baby details Patient Sticker Name Male/Female Date of Birth Hospital Number Address Baby Details DOA DOD Birth weight Gestational age Growth Type AGA SGA LGA Cord blood sample Arterial Venous Both Postnatal blood sample yes no Guthrie sample yes no Mode of delivery CS Instrumental Normal APGAR APGAR1 APGAR5 APGAR10 Resuscitation yes no ET Ventilation/CPAP yes no Sepsis yes no yes no Date of blood sampling Number of blood samples Medications TPN Age at feed intolerance Type of feed Volume of feed Method of feed % aspirate Colour of aspirate Days to full feeds Days to discharge WCC (differential count) Platelet count CRP 226 Chapter 14 – Appendices Blood culture Maternal Details Maternal Diagnosis Significant Antenatal/natal/postnatal history PROM yes no yes no Chorioamnionitis yes no Antenatal steroids yes no Medications 227 Chapter 15 – Bibliography Chapter 15- Bibliography Abaffy, T. and G. J. Cooper (2004). "GSK3 involvement in amylin signaling in isolated rat soleus muscle." Peptides 25(12): 2119-25. Abell, T. L., M. Camilleri, et al. (2008). "Consensus recommendations for gastric emptying scintigraphy: a joint report of the American Neurogastroenterology and Motility Society and the Society of Nuclear Medicine." J Nucl Med Technol 36(1): 44-54. Abell, T. L., M. Camilleri, et al. (2008). "Consensus recommendations for gastric emptying scintigraphy: a joint report of the American Neurogastroenterology and Motility Society and the Society of Nuclear Medicine." Am J Gastroenterol 103(3): 753-63. Abell, T. L. and J. R. Malagelada (1988). "Electrogastrography. Current assessment and future perspectives." Dig Dis Sci 33(8): 982-92. Ahronheim, J. (1943). "Nature of hyaline material in pancreatic islets in diabetes mellitus. ." Am J Pathol(19): 873-82. Akimoto, K., M. Nakazato, et al. (1993). "Plasma concentration of islet amyloid polypeptide in healthy children and patients with insulin-dependent diabetes mellitus." Acta Paediatr 82(3): 310-1. Akintorin, S. M., M. Kamat, et al. (1997). "A prospective randomized trial of feeding methods in very low birth weight infants." Pediatrics 100(4): E4. al Tawil, Y. and C. L. Berseth (1996). "Gestational and postnatal maturation of duodenal motor responses to intragastric feeding." J Pediatr 129(3): 374-81. Altman DG (1991). Practical statistics for medical research. London Chapman and Hall, . Amylin Pharmaceuticals (2003). Transplacental passage of amylin. 228 Chapter 15 – Bibliography Azriel, R. and E. Gazit (2001). "Analysis of the minimal amyloid-forming fragment of the islet amyloid polypeptide. An experimental support for the key role of the phenylalanine residue in amyloid formation." J Biol Chem 276(36): 34156-61. Badman, M. K., K. I. Shennan, et al. (1996). "Processing of pro-islet amyloid polypeptide (proIAPP) by the prohormone convertase PC2." FEBS Lett 378(3): 227-31. Baldo, B. A. and A. E. Kelley (2001). "Amylin infusion into rat nucleus accumbens potently depresses motor activity and ingestive behavior." Am J Physiol Regul Integr Comp Physiol 281(4): R1232-42. Banting, F. G. B., C.H.; Macleod, J.J.R (1922). "The internal secretion of the pancreas (Abstract). ." Am J Physiol 59: 479. Barnett, J. L. and C. Owyang (1988). "Serum glucose concentration as a modulator of interdigestive gastric motility." Gastroenterology 94(3): 739-44. Beardsall, K. and D. Dunger (2007). "The physiology and clinical management of glucose metabolism in the newborn." Endocr Dev 12: 124-37. Benini, L., C. Sembenini, et al. (1999). "Simultaneous measurement of gastric emptying of a solid meal by ultrasound and by scintigraphy." Am J Gastroenterol 94(10): 2861-5. Berseth, C. L. (1989). "Gestational evolution of small intestine motility in preterm and term infants." J Pediatr 115(4): 646-51. Berseth, C. L. (1990). "Neonatal small intestinal motility: motor responses to feeding in term and preterm infants." J Pediatr 117(5): 777-82. Berseth, C. L. (1996). "Gastrointestinal motility in the neonate." Clin Perinatol 23(2): 179-90. 229 Chapter 15 – Bibliography Berstad, A., T. Hausken, et al. (1996). "Ultrasonography of the human stomach." Scand J Gastroenterol Suppl 220: 75-82. Berthoug, H. N., WL (1993). Distribution and morphology of vagal afferents and efferents supplying the digestive system. In: Innervation of the Gut: Pathophysiological Implications. Innervation of the Gut: Pathophysiological Implications. Y. W. Tache, D; Burks, T. Boca Raton CRC Press. Bisset, W. M., J. B. Watt, et al. (1988). "Ontogeny of fasting small intestinal motor activity in the human infant." Gut 29(4): 483-8. Blair, E. H., R. R. Wing, et al. (1991). "The effect of laboratory stressors on glycemic control and gastrointestinal transit time." Psychosom Med 53(2): 13343. Bliss, M. (1982). "Banting's, Best's, and Collip's accounts of the discovery of insulin." Bull Hist Med 56(4): 554-68. Blumenthal, I., G. T. Lealman, et al. (1980). "Effect of feed temperature and phototherapy on gastric emptying in the neonate." Arch Dis Child 55(7): 562-4. Blumenthal, I. and R. S. Pildes (1979). "Effect of posture on the pattern of stomach emptying in the newborn." Pediatrics 63(4): 532-6. Bombell, S. and W. McGuire (2009). "Early trophic feeding for very low birth weight infants." Cochrane Database Syst Rev(3): CD000504. Boskovic, R., D. S. Feig, et al. (2003). "Transfer of insulin lispro across the human placenta: in vitro perfusion studies." Diabetes Care 26(5): 1390-4. Braegger, C., A. Chmielewska, et al. (2011). "Supplementation of infant formula with probiotics and/or prebiotics: a systematic review and comment by the ESPGHAN committee on nutrition." J Pediatr Gastroenterol Nutr 52(2): 238-50. Bronsky, J., M. Chada, et al. (2002). "[Amylin--its physiological role in humans]." Cesk Fysiol 51(4): 176-80. 230 Chapter 15 – Bibliography Bryant, M. G., A. M. Buchan, et al. (1982). "Development of intestinal regulatory peptides in the human fetus." Gastroenterology 83(1 Pt 1): 47-54. Butler, P. C., J. Chou, et al. (1990). "Effects of meal ingestion on plasma amylin concentration in NIDDM and nondiabetic humans." Diabetes 39(6): 752-6. Bytzer, P., N. J. Talley, et al. (2001). "Prevalence of gastrointestinal symptoms associated with diabetes mellitus: a population-based survey of 15,000 adults." Arch Intern Med 161(16): 1989-96. Cao, Z., P. J. Wookey, et al. (1997). "Renal amylin binding in normotensive and hypertensive rats: effects of angiotensin converting enzyme inhibition with perindopril." J Hypertens 15(11): 1245-52. Cavell, B. (1979). "Gastric emptying in preterm infants." Acta Paediatr Scand 68(5): 725-30. Cavell, B. (1981). "Gastric emptying in infants with congenital heart disease." Acta Paediatr Scand 70(4): 517-20. Cavell, B. (1982). "Reservoir and emptying function of the stomach of the premature infant." Acta Paediatr Scand Suppl 296: 60-1. Chaikomin, R., C. K. Rayner, et al. (2006). "Upper gastrointestinal function and glycemic control in diabetes mellitus." World J Gastroenterol 12(35): 5611-21. Challier, J. C., S. Hauguel, et al. (1986). "Effect of insulin on glucose uptake and metabolism in the human placenta." J Clin Endocrinol Metab 62(5): 803-7. Chen, J. D., E. Co, et al. (1997). "Patterns of gastric myoelectrical activity in human subjects of different ages." Am J Physiol 272(5 Pt 1): G1022-7. Clark, R. H., B. T. Bloom, et al. (2006). "Reported medication use in the neonatal intensive care unit: data from a large national data set." Pediatrics 117(6): 1979-87. 231 Chapter 15 – Bibliography Clementi, G., A. Caruso, et al. (1996). "Amylin given by central or peripheral routes decreases gastric emptying and intestinal transit in the rat." Experientia 52(7): 677-9. Clementi, G., A. Caruso, et al. (1995). "Anti-inflammatory activity of amylin and CGRP in different experimental models of inflammation." Life Sci 57(14): PL193-7. Clodi, M., K. Thomaseth, et al. (1998). "Distribution and kinetics of amylin in humans." Am J Physiol 274(5 Pt 1): E903-8. Collins, P. J., M. Horowitz, et al. (1983). "Gastric emptying in normal subjects--a reproducible technique using a single scintillation camera and computer system." Gut 24(12): 1117-25. Collins, P. J., L. A. Houghton, et al. (1991). "Role of the proximal and distal stomach in mixed solid and liquid meal emptying." Gut 32(6): 615-9. Combs, C. A., E. Gunderson, et al. (1992). "Relationship of fetal macrosomia to maternal postprandial glucose control during pregnancy." Diabetes Care 15(10): 1251-7. Cooke, R. J., S. B. Ainsworth, et al. (2004). "Postnatal growth retardation: a universal problem in preterm infants." Arch Dis Child Fetal Neonatal Ed 89(5): F428-30. Cooper, G. J. (1994). "Amylin compared with calcitonin gene-related peptide: structure, biology, and relevance to metabolic disease." Endocr Rev 15(2): 163201. Cooper, G. J., A. J. Day, et al. (1989). "Amylin and the amylin gene: structure, function and relationship to islet amyloid and to diabetes mellitus." Biochim Biophys Acta 1014(3): 247-58. 232 Chapter 15 – Bibliography Cooper, G. J., B. Leighton, et al. (1988). "Amylin found in amyloid deposits in human type 2 diabetes mellitus may be a hormone that regulates glycogen metabolism in skeletal muscle." Proc Natl Acad Sci U S A 85(20): 7763-6. Cooper, G. J., A. C. Willis, et al. (1987). "Purification and characterization of a peptide from amyloid-rich pancreases of type 2 diabetic patients." Proc Natl Acad Sci U S A 84(23): 8628-32. Cooper, M. E., P. G. McNally, et al. (1995). "Amylin stimulates plasma renin concentration in humans." Hypertension 26(3): 460-4. Cornish, J., K. E. Callon, et al. (2001). "Effects of calcitonin, amylin, and calcitonin gene-related peptide on osteoclast development." Bone 29(2): 162-8. Cornish, J., K. E. Callon, et al. (1995). "Amylin stimulates osteoblast proliferation and increases mineralized bone volume in adult mice." Biochem Biophys Res Commun 207(1): 133-9. Cornish, J., K. E. Callon, et al. (1998). "Dissociation of the effects of amylin on osteoblast proliferation and bone resorption." Am J Physiol 274(5 Pt 1): E82733. Cornish, J. and D. Naot (2002). "Amylin and adrenomedullin: novel regulators of bone growth." Curr Pharm Des 8(23): 2009-21. Costalos, C., I. Ross, et al. (1979). "Is it necessary to warm infants' feeds?" Arch Dis Child 54(11): 899-901. Costalos, C., G. Russell, et al. (1984). "Effects of jaundice and phototherapy on gastric emptying in the newborn." Biol Neonate 46(2): 57-60. Crowther, C. A., J. E. Hiller, et al. (2005). "Effect of treatment of gestational diabetes mellitus on pregnancy outcomes." N Engl J Med 352(24): 2477-86. Crowther, J. R. (1995). Methods in Molecular Biology. ELISA, Humana Press. Volume 42: 35-61. 233 Chapter 15 – Bibliography Currao, W. J., C. Cox, et al. (1988). "Diluted formula for beginning the feeding of premature infants." Am J Dis Child 142(7): 730-1. Daniel, E. E. and Y. F. Wang (1999). "Control systems of gastrointestinal motility are immature at birth in dogs." Neurogastroenterol Motil 11(5): 375-92. Delmis, J., A. Drazancic, et al. (1992). "Glucose, insulin, HGH and IGF-I levels in maternal serum, amniotic fluid and umbilical venous serum: a comparison between late normal pregnancy and pregnancies complicated with diabetes and fetal growth retardation." J Perinat Med 20(1): 47-56. Deshpande, G., S. Rao, et al. (2007). "Probiotics for prevention of necrotising enterocolitis in preterm neonates with very low birthweight: a systematic review of randomised controlled trials." Lancet 369(9573): 1614-20. Deshpande, G., S. Rao, et al. (2010). "Updated meta-analysis of probiotics for preventing necrotizing enterocolitis in preterm neonates." Pediatrics 125(5): 921-30. Directive, E. (2008). "Recommendations of the ad hoc group for the development of implementing guidelines for Directive 2001/20/EC relating to good clinical practice in the conduct of clinical trials on medicinal products for human use. Ethical considerations for clinical trials on medicinal products conducted with the paediatric population. 2008.". Dollberg, S., J. Kuint, et al. (2000). "Feeding tolerance in preterm infants: randomized trial of bolus and continuous feeding." J Am Coll Nutr 19(6): 797800. Donnell, S. C., N. Taylor, et al. (2002). "Infection rates in surgical neonates and infants receiving parenteral nutrition: a five-year prospective study." J Hosp Infect 52(4): 273-80. Druce, M. and S. R. Bloom (2006). "The regulation of appetite." Arch Dis Child 91(2): 183-7. 234 Chapter 15 – Bibliography Dunn, L., S. Hulman, et al. (1988). "Beneficial effects of early hypocaloric enteral feeding on neonatal gastrointestinal function: preliminary report of a randomized trial." J Pediatr 112(4): 622-9. Dyer, J. S., C. R. Rosenfeld, et al. (2007). "Insulin resistance in Hispanic largefor-gestational-age neonates at birth." J Clin Endocrinol Metab 92(10): 3836-43. Economides, D. L., K. H. Nicolaides, et al. (1990). "Relation between maternalto-fetal blood glucose gradient and uterine and umbilical Doppler blood flow measurements." Br J Obstet Gynaecol 97(6): 543-4. Edelman, S. V. and C. Weyer (2002). "Unresolved challenges with insulin therapy in type 1 and type 2 diabetes: potential benefit of replacing amylin, a second beta-cell hormone." Diabetes Technol Ther 4(2): 175-89. Edwards, G. G., BR; Jodka, C; Dilts, RP; Miller, CC; Young, A (1998). "Area postrema (AP)-lesions block the regulation of gastric emptying by amylin (Abstract)." Neurogastroenterol Motil 10(26A). Ehrenkranz, R. A., N. Younes, et al. (1999). "Longitudinal growth of hospitalized very low birth weight infants." Pediatrics 104(2 Pt 1): 280-9. Ehrlich, J. and I. Ratner (1961). "Amyloidosis of the islets of Langerhans. A restudy of islet hyaline in diabetic and non diabetic individuals." Am J Pathol(38): 49-59. ElHennawy, A. A., J. W. Sparks, et al. (2003). "Erythromycin fails to improve feeding outcome in feeding-intolerant preterm infants." J Pediatr Gastroenterol Nutr 37(3): 281-6. Ernst, J. A., K. A. Rickard, et al. (1989). "Lack of improved growth outcome related to nonnutritive sucking in very low birth weight premature infants fed a controlled nutrient intake: a randomized prospective study." Pediatrics 83(5): 706-16. 235 Chapter 15 – Bibliography Ewer, A. K., G. M. Durbin, et al. (1994). "Gastric emptying in preterm infants." Arch Dis Child Fetal Neonatal Ed 71(1): F24-7. Familoni, B. O., K. L. Bowes, et al. (1991). "Can transcutaneous recordings detect gastric electrical abnormalities?" Gut 32(2): 141-6. Fan, L., G. Westermark, et al. (1994). "Altered gene structure and tissue expression of islet amyloid polypeptide in the chicken." Mol Endocrinol 8(6): 713-21. Feinle, C., P. Kunz, et al. (1999). "Scintigraphic validation of a magnetic resonance imaging method to study gastric emptying of a solid meal in humans." Gut 44(1): 106-11. Ferrier, G. J., A. M. Pierson, et al. (1989). "Expression of the rat amylin (IAPP/DAP) gene." J Mol Endocrinol 3(1): R1-4. Fineman, M., C. Weyer, et al. (2002). "The human amylin analog, pramlintide, reduces postprandial hyperglucagonemia in patients with type 2 diabetes mellitus." Horm Metab Res 34(9): 504-8. Fineman MS, G. M., Thompson RG, Kolterman OG, Koda JE (1996). "Amylin response following Sustacal® ingestion is diminished in type II diabetic patients treated with insulin (Abstract)." Diabetologia 39 (Suppl. 1): A149. Fowden, A. L. (1989). "The role of insulin in prenatal growth." J Dev Physiol 12(4): 173-82. Frank, L. and I. R. Sosenko (1988). "Undernutrition as a major contributing factor in the pathogenesis of bronchopulmonary dysplasia." Am Rev Respir Dis 138(3): 725-9. Frost, G. S., A. E. Brynes, et al. (2003). "The effects of fiber enrichment of pasta and fat content on gastric emptying, GLP-1, glucose, and insulin responses to a meal." Eur J Clin Nutr 57(2): 293-8. 236 Chapter 15 – Bibliography Garland, S. M., J. M. Tobin, et al. (2011). "The ProPrems trial: investigating the effects of probiotics on late onset sepsis in very preterm infants." BMC Infect Dis 11: 210. Gedulin, B. J., CM; Greene, DL; Young AA (1996). Comparison of 21 peptides on inhibition of gastric emptying in conscious rats (Abstract). Digestive Disease Week, American \association for the Study of Liver Disease, American Gastroenterological Association: A-742. Gedulin, B. R. and A. A. Young (1998). "Hypoglycemia overrides amylinmediated regulation of gastric emptying in rats." Diabetes 47(1): 93-7. Gentilcore, D., T. Hausken, et al. (2006). "Measurements of gastric emptying of low- and high-nutrient liquids using 3D ultrasonography and scintigraphy in healthy subjects." Neurogastroenterol Motil 18(12): 1062-8. George, J. D. (1968). "New clinical method for measuring the rate of gastric emptying: the double sampling test meal." Gut 9(2): 237-42. Geraldo V, P. S., Joseph T, Pildes RS. (1997). "Gastric residual (GR): reliability of the measurement." Pediatr Res. 41: 150A. Gesteiro, E., S. Bastida, et al. (2009). "Insulin resistance markers in term, normoweight neonates. The Merida cohort." Eur J Pediatr 168(3): 281-8. Gillman, M. W., H. Oakey, et al. (2010). "Effect of treatment of gestational diabetes mellitus on obesity in the next generation." Diabetes Care 33(5): 9648. Golpon, H. A., A. Puechner, et al. (2001). "Vasorelaxant effect of glucagon-like peptide-(7-36)amide and amylin on the pulmonary circulation of the rat." Regul Pept 102(2-3): 81-6. Gomes, H., P. Hornoy, et al. (2003). "Ultrasonography and gastric emptying in children: validation of a sonographic method and determination of physiological and pathological patterns." Pediatr Radiol 33(8): 522-9. 237 Chapter 15 – Bibliography Gounaris, A., C. Costalos, et al. (2010). "Gastric emptying of preterm neonates receiving domperidone." Neonatology 97(1): 56-60. Green, J. D., C. Goldsbury, et al. (2004). "Human amylin oligomer growth and fibril elongation define two distinct phases in amyloid formation." J Biol Chem 279(13): 12206-12. Gross, P. (1992). Circumventricular organ capillaries. In: Circumventricular Organs and Brain Fluid Environment. Amsterdam, The Netherlands, Elsevier. Gross, S. J. (1983). "Growth and biochemical response of preterm infants fed human milk or modified infant formula." N Engl J Med 308(5): 237-41. Gross, S. J. and T. A. Slagle (1993). "Feeding the low birth weight infant." Clin Perinatol 20(1): 193-209. Grover, A., M. Khashu, et al. (2008). "Iatrogenic malnutrition in neonatal intensive care units: urgent need to modify practice." JPEN J Parenter Enteral Nutr 32(2): 140-4. Gupta, M. and Y. W. Brans (1978). "Gastric retention in neonates." Pediatrics 62(1): 26-9. Hack, M., N. Breslau, et al. (1991). "Effect of very low birth weight and subnormal head size on cognitive abilities at school age." N Engl J Med 325(4): 231-7. Hack, M., M. Schluchter, et al. (2003). "Growth of very low birth weight infants to age 20 years." Pediatrics 112(1 Pt 1): e30-8. Hall, J. M. and S. D. Brain (1999). "Interaction of amylin with calcitonin generelated peptide receptors in the microvasculature of the hamster cheek pouch in vivo." Br J Pharmacol 126(1): 280-4. 238 Chapter 15 – Bibliography Harris, P. J., M. E. Cooper, et al. (1997). "Amylin stimulates proximal tubular sodium transport and cell proliferation in the rat kidney." Am J Physiol 272(1 Pt 2): F13-21. Hartter, E., T. Svoboda, et al. (1991). "Basal and stimulated plasma levels of pancreatic amylin indicate its co-secretion with insulin in humans." Diabetologia 34(1): 52-4. Hawdon, J. M., A. Aynsley-Green, et al. (1993). "The role of pancreatic insulin secretion in neonatal glucoregulation. I. Healthy term and preterm infants." Arch Dis Child 68(3 Spec No): 274-9. Hay, D. L., G. Christopoulos, et al. (2004). "Amylin receptors: molecular composition and pharmacology." Biochem Soc Trans 32(Pt 5): 865-7. Hay, W. W., Jr. (2008). "Strategies for feeding the preterm infant." Neonatology 94(4): 245-54. Hayden, M. R. and S. C. Tyagi (2001). ""A" is for amylin and amyloid in type 2 diabetes mellitus." JOP 2(4): 124-39. Haynes, J. M., W. C. Hodgson, et al. (1997). "Rat amylin mediates a pressor response in the anaesthetised rat: implications for the association between hypertension and diabetes mellitus." Diabetologia 40(3): 256-61. Heading, R. C., J. Nimmo, et al. (1973). "The dependence of paracetamol absorption on the rate of gastric emptying." Br J Pharmacol 47(2): 415-21. Hellstrom, P. M., P. Gryback, et al. (2006). "The physiology of gastric emptying." Best Pract Res Clin Anaesthesiol 20(3): 397-407. Hemachandra, A. H. and R. M. Cowett (1999). "Neonatal hyperglycemia." Pediatrics in Review Vol. 20: e16 -e24. 239 Chapter 15 – Bibliography Heptulla, R. A., L. M. Rodriguez, et al. (2008). "Gastric emptying and postprandial glucose excursions in adolescents with type 1 diabetes." Pediatr Diabetes 9(6): 561-6. Heyland, D. K., G. Tougas, et al. (1996). "Cisapride improves gastric emptying in mechanically ventilated, critically ill patients. A randomized, double-blind trial." Am J Respir Crit Care Med 154(6 Pt 1): 1678-83. Heyland, D. K., G. Tougas, et al. (1996). "Impaired gastric emptying in mechanically ventilated, critically ill patients." Intensive Care Med 22(12): 133944. Hibbs, A. M. and S. A. Lorch (2006). "Metoclopramide for the treatment of gastroesophageal reflux disease in infants: a systematic review." Pediatrics 118(2): 746-52. Hoffman, L., C. Nolan, et al. (1998). "Gestational diabetes mellitus-management guidelines. The Australasian Diabetes in Pregnancy Society." Med J Aust 169(2): 93-7. Hollander, P. A., P. Levy, et al. (2003). "Pramlintide as an adjunct to insulin therapy improves long-term glycemic and weight control in patients with type 2 diabetes: a 1-year randomized controlled trial." Diabetes Care 26(3): 784-90. Holt, S., J. Cervantes, et al. (1986). "Measurement of gastric emptying rate in humans by real-time ultrasound." Gastroenterology 90(4): 918-23. Hoppener, J. W., B. Ahren, et al. (2000). "Islet amyloid and type 2 diabetes mellitus." N Engl J Med 343(6): 411-9. Horowitz, M., D. O'Donovan, et al. (2002). "Gastric emptying in diabetes: clinical significance and treatment." Diabet Med 19(3): 177-94. Hossain, P., B. Kawar, et al. (2007). "Obesity and diabetes in the developing world--a growing challenge." N Engl J Med 356(3): 213-5. 240 Chapter 15 – Bibliography Hunt, J. N. and M. T. Knox (1968). "Control of gastric emptying." Am J Dig Dis 13(4): 372-5. Hunt, L. I., D. L. Antonson, et al. (1982). "Osmolality of carbohydrate solutions and gastric emptying in the newborn." Am J Dis Child 136(5): 448-51. Husband, J. and P. Husband (1969). "Gastric emptying of water and glucose solutions in the newborn." Lancet 2(7617): 409-11. Husband, J., P. Husband, et al. (1970). "Gastric emptying of starch meals in the newborn." Lancet 2(7667): 290-2. Ikeda, T., K. Iwata, et al. (2001). "Effect of insulin, proinsulin, and amylin on renin release from perfused rat kidney." Metabolism 50(7): 763-6. Ittmann, P. I., R. Amarnath, et al. (1992). "Maturation of antroduodenal motor activity in preterm and term infants." Dig Dis Sci 37(1): 14-9. Jadcherla, S. R. and R. M. Kliegman (2002). "Studies of feeding intolerance in very low birth weight infants: definition and significance." Pediatrics 109(3): 5167. Jaikaran, E. T. and A. Clark (2001). "Islet amyloid and type 2 diabetes: from molecular misfolding to islet pathophysiology." Biochim Biophys Acta 1537(3): 179-203. Jaikaran, E. T., C. E. Higham, et al. (2001). "Identification of a novel human islet amyloid polypeptide beta-sheet domain and factors influencing fibrillogenesis." J Mol Biol 308(3): 515-25. Jayasinghe, S. A. and R. Langen (2004). "Identifying structural features of fibrillar islet amyloid polypeptide using site-directed spin labeling." J Biol Chem 279(46): 48420-5. 241 Chapter 15 – Bibliography Jodka, C. G., D; Young, A; Gedulin, B (1996). "Amylin modulation of gastric emptying in rats depends upon an intact vagus nerve (Abstract)." Diabetes 45 (Suppl. 2)(235A). Jovanovic-Peterson, L., C. M. Peterson, et al. (1991). "Maternal postprandial glucose levels and infant birth weight: the Diabetes in Early Pregnancy Study. The National Institute of Child Health and Human Development--Diabetes in Early Pregnancy Study." Am J Obstet Gynecol 164(1 Pt 1): 103-11. Joyce, C. D., R. R. Fiscus, et al. (1990). "Calcitonin gene-related peptide levels are elevated in patients with sepsis." Surgery 108(6): 1097-101. Juhl, C. B., N. Porksen, et al. (2000). "High-frequency oscillations in circulating amylin concentrations in healthy humans." Am J Physiol Endocrinol Metab 278(3): E484-90. Kairamkonda, V., A. Deorukhkar, et al. (2005). "Amylin peptide levels are raised in infants of diabetic mothers." Arch Dis Child 90(12): 1279-82. Kajava, A. V., U. Aebi, et al. (2005). "The parallel superpleated beta-structure as a model for amyloid fibrils of human amylin." J Mol Biol 348(2): 247-52. Karlsson, E. and S. Sandler (2001). "Islet amyloid polypeptide promotes betacell proliferation in neonatal rat pancreatic islets." Diabetologia 44(8): 1015-8. Kaufman, S. S., G. E. Gondolesi, et al. (2003). "Parenteral nutrition associated liver disease." Semin Neonatol 8(5): 375-81. Keller, J. M. and J. S. Krohmer (1968). "Insulin transfer in the isolated human placenta." Obstet Gynecol 32(1): 77-80. Kelley, D., A. Mitrakou, et al. (1988). "Skeletal muscle glycolysis, oxidation, and storage of an oral glucose load." J Clin Invest 81(5): 1563-71. Kellmeyer, T. A., N. C. Kesty, et al. (2007). "Pharmacokinetics of an oral drug (acetaminophen) administered at various times relative to subcutaneous 242 Chapter 15 – Bibliography injection of pramlintide in subjects with type 2 diabetes." J Clin Pharmacol 47(7): 798-805. Kim, D. Y., S. J. Myung, et al. (2000). "Novel testing of human gastric motor and sensory functions: rationale, methods, and potential applications in clinical practice." Am J Gastroenterol 95(12): 3365-73. Kitzmiller, J. L., J. P. Cloherty, et al. (1978). "Diabetic pregnancy and perinatal morbidity." Am J Obstet Gynecol 131(5): 560-80. Kliegman, R. M. (1999). "Experimental validation of neonatal feeding practices." Pediatrics 103(2): 492-3. Knowler, W. C., E. Barrett-Connor, et al. (2002). "Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin." N Engl J Med 346(6): 393-403. Knowler, W. C., S. E. Fowler, et al. (2009). "10-year follow-up of diabetes incidence and weight loss in the Diabetes Prevention Program Outcomes Study." Lancet 374(9702): 1677-86. Koda, J. E., M. Fineman, et al. (1992). "Amylin concentrations and glucose control." Lancet 339(8802): 1179-80. Koda JE, F. M., Kolterman OG, and C. JF (1995). "24 hour plasma amylin profiles are elevated in IGT subjects vs. normal controls (Abstract)." Diabetes 44 (Suppl. 1): 238A. Kojima, M., H. Hosoda, et al. (1999). "Ghrelin is a growth-hormone-releasing acylated peptide from stomach." Nature 402(6762): 656-60. Kolterman, O. G., A. Gottlieb, et al. (1995). "Reduction of postprandial hyperglycemia in subjects with IDDM by intravenous infusion of AC137, a human amylin analogue." Diabetes Care 18(8): 1179-82. 243 Chapter 15 – Bibliography Kong, M. F., P. King, et al. (1999). "Euglycaemic hyperinsulinaemia does not affect gastric emptying in type I and type II diabetes mellitus." Diabetologia 42(3): 365-72. Kong, M. F., P. King, et al. (1997). "Infusion of pramlintide, a human amylin analogue, delays gastric emptying in men with IDDM." Diabetologia 40(1): 82-8. Kong, M. F., T. A. Stubbs, et al. (1998). "The effect of single doses of pramlintide on gastric emptying of two meals in men with IDDM." Diabetologia 41(5): 577-83. Kuo, B., R. W. McCallum, et al. (2008). "Comparison of gastric emptying of a nondigestible capsule to a radio-labelled meal in healthy and gastroparetic subjects." Aliment Pharmacol Ther 27(2): 186-96. Kurganov, B., M. Doh, et al. (2004). "Aggregation of liposomes induced by the toxic peptides Alzheimer's Abetas, human amylin and prion (106-126): facilitation by membrane-bound GM1 ganglioside." Peptides 25(2): 217-32. Lain, K. Y., M. J. Garabedian, et al. (2009). "Neonatal adiposity following maternal treatment of gestational diabetes with glyburide compared with insulin." Am J Obstet Gynecol 200(5): 501 e1-6. Landon, M. B., C. Y. Spong, et al. (2009). "A multicenter, randomized trial of treatment for mild gestational diabetes." N Engl J Med 361(14): 1339-48. Langerhans, P. (1869). Beiträge zur mikroskopischen Anatomie der Bauchspeicheldrüse. Inaugural-Dissertation. Berlin:Gustav Lange. Languesse, E. (1893). "Sur la formation des lots de Langerhans dans le pancreas." Comptes Rend Soc Biol 5 (series 9): 819-820. Lee-Parritz, C. J. (2004). Maternal conditions that affect the fetus: diabetes mellitus. Manual of Neonatal Care (Fifth Edition). E. E. Cloherty JP, Stark AR. Philadelphia, PA Lippincott Williams & Wilkins: 9-19. 244 Chapter 15 – Bibliography Liang, J., E. Co, et al. (1998). "Development of gastric slow waves in preterm infants measured by electrogastrography." Am J Physiol 274(3 Pt 1): G503-8. Lucas, A., D. L. Sarson, et al. (1980). "Breast vs bottle: endocrine responses are different with formula feeding." Lancet 1(8181): 1267-9. Ludvik, B., A. Kautzky-Willer, et al. (1997). "Amylin: history and overview." Diabet Med 14 Suppl 2: S9-13. Ludvik, B., B. Lell, et al. (1991). "Decrease of stimulated amylin release precedes impairment of insulin secretion in type II diabetes." Diabetes 40(12): 1615-9. Ludvik, B., K. Thomaseth, et al. (2003). "Inverse relation between amylin and glucagon secretion in healthy and diabetic human subjects." Eur J Clin Invest 33(4): 316-22. Luk'yantseva, G. V., I. Y. Sergeev, et al. (2001). "Effect of amylin on the tone of rat aorta ring preparation." Bull Exp Biol Med 132(4): 932-4. Lutz, T. A., E. Del Prete, et al. (1994). "Reduction of food intake in rats by intraperitoneal injection of low doses of amylin." Physiol Behav 55(5): 891-5. Lutz, T. A., E. Del Prete, et al. (1995). "Subdiaphragmatic vagotomy does not influence the anorectic effect of amylin." Peptides 16(3): 457-62. Lutz, T. A., N. Geary, et al. (1995). "Amylin decreases meal size in rats." Physiol Behav 58(6): 1197-202. Lutz, T. A., A. Mollet, et al. (2001). "The anorectic effect of a chronic peripheral infusion of amylin is abolished in area postrema/nucleus of the solitary tract (AP/NTS) lesioned rats." Int J Obes Relat Metab Disord 25(7): 1005-11. Lutz, T. A., M. Senn, et al. (1998). "Lesion of the area postrema/nucleus of the solitary tract (AP/NTS) attenuates the anorectic effects of amylin and calcitonin gene-related peptide (CGRP) in rats." Peptides 19(2): 309-17. 245 Chapter 15 – Bibliography Lutz, T. A., S. Tschudy, et al. (2001). "Dopamine D(2) receptors mediate amylin's acute satiety effect." Am J Physiol Regul Integr Comp Physiol 280(6): R1697-703. Macdonald, I. A. (1997). "Amylin and the gastrointestinal tract." Diabet Med 14 Suppl 2: S24-8. Maclennan, S., C. Augood, et al. (2010). "Cisapride treatment for gastrooesophageal reflux in children." Cochrane Database Syst Rev 4: CD002300. Mayer, A. P., A. Durward, et al. (2002). "Amylin is associated with delayed gastric emptying in critically ill children." Intensive Care Med 28(3): 336-40. McGuire, W. and P. McEwan (2007). "Transpyloric versus gastric tube feeding for preterm infants." Cochrane Database Syst Rev(3): CD003487. Meetze, W. H., C. Valentine, et al. (1992). "Gastrointestinal priming prior to full enteral nutrition in very low birth weight infants." J Pediatr Gastroenterol Nutr 15(2): 163-70. Mericq, V. (2006). "Low birth weight and endocrine dysfunction in postnatal life." Pediatr Endocrinol Rev 4(1): 3-14. Mericq, V. (2006). "Prematurity and insulin sensitivity." Horm Res 65 Suppl 3: 131-6. Metzger, B. E. and D. R. Coustan (1998). "Summary and recommendations of the Fourth International Workshop-Conference on Gestational Diabetes Mellitus. The Organizing Committee." Diabetes Care 21 Suppl 2: B161-7. Meyer, C., J. M. Dostou, et al. (2002). "Role of human liver, kidney, and skeletal muscle in postprandial glucose homeostasis." Am J Physiol Endocrinol Metab 282(2): E419-27. 246 Chapter 15 – Bibliography Mihatsch, W. A., P. von Schoenaich, et al. (2002). "The significance of gastric residuals in the early enteral feeding advancement of extremely low birth weight infants." Pediatrics 109(3): 457-9. Mintz, G. R. (1993). "An irreversible serine protease inhibitor." BioPharm(6): 3438. Mitanchez-Mokhtari, D., N. Lahlou, et al. (2004). "Both relative insulin resistance and defective islet beta-cell processing of proinsulin are responsible for transient hyperglycemia in extremely preterm infants." Pediatrics 113(3 Pt 1): 537-41. Mitsukawa, T., J. Takemura, et al. (1990). "Islet amyloid polypeptide response to glucose, insulin, and somatostatin analogue administration." Diabetes 39(5): 639-42. Mok, K. H., J. Pettersson, et al. (2007). "HAMLET, protein folding, and tumor cell death." Biochem Biophys Res Commun 354(1): 1-7. Mollet, A., S. Gilg, et al. (2004). "Infusion of the amylin antagonist AC 187 into the area postrema increases food intake in rats." Physiol Behav 81(1): 149-55. Mollet, A., T. A. Lutz, et al. (2001). "Histamine H1 receptors mediate the anorectic action of the pancreatic hormone amylin." Am J Physiol Regul Integr Comp Physiol 281(5): R1442-8. Moore, J. G., P. E. Christian, et al. (1981). "Gastric emptying of varying meal weight and composition in man. Evaluation by dual liquid- and solid-phase isotopic method." Dig Dis Sci 26(1): 16-22. Morley, J. E. and J. F. Flood (1991). "Amylin decreases food intake in mice." Peptides 12(4): 865-9. Morley, J. E. and J. F. Flood (1994). "Effect of competitive antagonism of NO synthetase on weight and food intake in obese and diabetic mice." Am J Physiol 266(1 Pt 2): R164-8. 247 Chapter 15 – Bibliography Morley, J. E., J. F. Flood, et al. (1995). "Effects of amylin on appetite regulation and memory." Can J Physiol Pharmacol 73(7): 1042-6. Morris, M. J. and T. Nguyen (2001). "Does neuropeptide Y contribute to the anorectic action of amylin?" Peptides 22(3): 541-6. Morrison, H. (1937). "Contribution to the Microscopic Anatomy of the Pancreas 1937 By Paul Langerhans (Berlin, 1869). Reprint of the German original with an English translation and an introductory essay." Bull Inst Hist Med 5: 259-297. Moses, R. G., M. Barker, et al. (2009). "Can a low-glycemic index diet reduce the need for insulin in gestational diabetes mellitus? A randomized trial." Diabetes Care 32(6): 996-1000. Moses, R. G., M. Luebcke, et al. (2006). "Effect of a low-glycemic-index diet during pregnancy on obstetric outcomes." Am J Clin Nutr 84(4): 807-12. Mulder, H., M. Ekelund, et al. (1997). "Islet amyloid polypeptide in the gut and pancreas: localization, ontogeny and gut motility effects." Peptides 18(6): 77183. Mulder, H., A. Leckstrom, et al. (1995). "Islet amyloid polypeptide (amylin) is expressed in sensory neurons." J Neurosci 15(11): 7625-32. Mulder, H., A. C. Lindh, et al. (1994). "Islet amyloid polypeptide is expressed in endocrine cells of the gastric mucosa in the rat and mouse." Gastroenterology 107(3): 712-9. Murphy, H. R., G. Rayman, et al. (2008). "Effectiveness of continuous glucose monitoring in pregnant women with diabetes: randomised clinical trial." BMJ 337: a1680. Nagata, S., T. Koyanagi, et al. (1994). "Change in the three-dimensional shape of the stomach in the developing human fetus." Early Hum Dev 37(1): 27-38. 248 Chapter 15 – Bibliography Ng, E. and V. S. Shah (2008). "Erythromycin for the prevention and treatment of feeding intolerance in preterm infants." Cochrane Database Syst Rev(3): CD001815. Ng, P. C., K. W. So, et al. (2001). "Randomised controlled study of oral erythromycin for treatment of gastrointestinal dysmotility in preterm infants." Arch Dis Child Fetal Neonatal Ed 84(3): F177-82. Nicholson, W., S. Bolen, et al. (2009). "Benefits and risks of oral diabetes agents compared with insulin in women with gestational diabetes: a systematic review." Obstet Gynecol 113(1): 193-205. Nilsson, M. R., M. Driscoll, et al. (2002). "Low levels of asparagine deamidation can have a dramatic effect on aggregation of amyloidogenic peptides: implications for the study of amyloid formation." Protein Sci 11(2): 342-9. Nishi, M., T. Sanke, et al. (1989). "Human islet amyloid polypeptide gene: complete nucleotide sequence, chromosomal localization, and evolutionary history." Mol Endocrinol 3(11): 1775-81. Nold, J. L. and M. K. Georgieff (2004). "Infants of diabetic mothers." Pediatr Clin North Am 51(3): 619-37, viii. Ochiai, T., Y. Chijiiwa, et al. (2001). "Direct inhibitory effect of adrenomedullin, calcitonin gene-related peptide, calcitonin, and amylin on cholecystokinininduced contraction of guinea-pig isolated caecal circular smooth muscle cells." Peptides 22(6): 909-14. Omari, T. I., N. Rommel, et al. (2004). "Paradoxical impact of body positioning on gastroesophageal reflux and gastric emptying in the premature neonate." J Pediatr 145(2): 194-200. Ong, K., J. Kratzsch, et al. (2000). "Size at birth and cord blood levels of insulin, insulin-like growth factor I (IGF-I), IGF-II, IGF-binding protein-1 (IGFBP-1), IGFBP-3, and the soluble IGF-II/mannose-6-phosphate receptor in term human 249 Chapter 15 – Bibliography infants. The ALSPAC Study Team. Avon Longitudinal Study of Pregnancy and Childhood." J Clin Endocrinol Metab 85(11): 4266-9. Onuoha, G. N. and E. K. Alpar (1999). "Calcitonin gene-related peptide and other neuropeptides in the plasma of patients with soft tissue injury." Life Sci 65(13): 1351-8. Opie, E. L. (1901). "The relation of diabetes mellitus to lesions of the pancreas: hyaline degeneration of the islands of Langerhans " J Exp Med(5): 527-40. Padrick, S. B. and A. D. Miranker (2001). "Islet amyloid polypeptide: identification of long-range contacts and local order on the fibrillogenesis pathway." J Mol Biol 308(4): 783-94. Parida, S. K., D. B. Schneider, et al. (1998). "Elevated circulating calcitonin gene-related peptide in umbilical cord and infant blood associated with maternal and neonatal sepsis and shock." Pediatr Res 43(2): 276-82. Parkman, H. P., W. L. Hasler, et al. (2004). "American Gastroenterological Association technical review on the diagnosis and treatment of gastroparesis." Gastroenterology 127(5): 1592-622. Patole, S. (2005). "Strategies for prevention of feed intolerance in preterm neonates: a systematic review." J Matern Fetal Neonatal Med 18(1): 67-76. Patole, S., S. Rao, et al. (2005). "Erythromycin as a prokinetic agent in preterm neonates: a systematic review." Arch Dis Child Fetal Neonatal Ed 90(4): F3016. Pedersen, J. (1977). The pregnant diabetic and her newborn. . Baltimore, Williams and Wilkins. Petitt, D. J., P. H. Bennett, et al. (1985). "Gestational diabetes mellitus and impaired glucose tolerance during pregnancy. Long-term effects on obesity and glucose tolerance in the offspring." Diabetes 34 Suppl 2: 119-22. 250 Chapter 15 – Bibliography Pettersson-Kastberg, J., S. Aits, et al. (2008). "Can misfolded proteins be beneficial? The HAMLET case." Ann Med: 1-15. Pettitt, D. J., K. A. Aleck, et al. (1988). "Congenital susceptibility to NIDDM. Role of intrauterine environment." Diabetes 37(5): 622-8. Phillips, A. R., F. M. Abu-Zidan, et al. (1999). "Intestinal ischaemia-reperfusion increases plasma amylin concentration in rats." Eur Surg Res 31(6): 457-64. Phillips, A. R., F. M. Abu-Zidan, et al. (2001). "Plasma amylin concentration is related to the severity of intestinal ischemic injury in rats." Surgery 129(6): 7305. Piao, F. L., C. Cao, et al. (2004). "Amylin-induced suppression of ANP secretion through receptors for CGRP1 and salmon calcitonin." Regul Pept 117(3): 15966. Poyner, D. R., P. M. Sexton, et al. (2002). "International Union of Pharmacology. XXXII. The mammalian calcitonin gene-related peptides, adrenomedullin, amylin, and calcitonin receptors." Pharmacol Rev 54(2): 23346. Pozler, O., D. Neumann, et al. (2003). "Development of gastric emptying in premature infants. Use of the (13)C-octanoic acid breath test." Nutrition 19(7-8): 593-6. Premji, S. S., J. Wilson, et al. (1997). "Cisapride: a review of the evidence supporting its use in premature infants with feeding intolerance." Neonatal Netw 16(7): 17-21. Putnam, P. E., S. R. Orenstein, et al. (1992). "Tardive dyskinesia associated with use of metoclopramide in a child." J Pediatr 121(6): 983-5. Ramirez, A., W. W. Wong, et al. (2006). "Factors regulating gastric emptying in preterm infants." J Pediatr 149(4): 475-9. 251 Chapter 15 – Bibliography Ratner, R. E., C. A. Christophi, et al. (2008). "Prevention of diabetes in women with a history of gestational diabetes: effects of metformin and lifestyle interventions." J Clin Endocrinol Metab 93(12): 4774-9. Ratner, R. E., L. L. Want, et al. (2002). "Adjunctive therapy with the amylin analogue pramlintide leads to a combined improvement in glycemic and weight control in insulin-treated subjects with type 2 diabetes." Diabetes Technol Ther 4(1): 51-61. Rayner, C. K., M. Samsom, et al. (2001). "Relationships of upper gastrointestinal motor and sensory function with glycemic control." Diabetes Care 24(2): 371-81. Rayyis, S. F., N. Ambalavanan, et al. (1999). "Randomized trial of "slow" versus "fast" feed advancements on the incidence of necrotizing enterocolitis in very low birth weight infants." J Pediatr 134(3): 293-7. Reda, T. K., A. Geliebter, et al. (2002). "Amylin, food intake, and obesity." Obes Res 10(10): 1087-91. Reddy, P. S., A. K. Deorari, et al. (2000). "A double-blind placebo-controlled study on prophylactic use of cisapride on feed intolerance and gastric emptying in preterm neonates." Indian Pediatr 37(8): 837-44. Riddle, M., J. Frias, et al. (2007). "Pramlintide improved glycemic control and reduced weight in patients with type 2 diabetes using basal insulin." Diabetes Care 30(11): 2794-9. Riediger, T., M. Rauch, et al. (1999). "Actions of amylin on subfornical organ neurons and on drinking behavior in rats." Am J Physiol 276(2 Pt 2): R514-21. Riediger, T., H. A. Schmid, et al. (2001). "Amylin potently activates AP neurons possibly via formation of the excitatory second messenger cGMP." Am J Physiol Regul Integr Comp Physiol 281(6): R1833-43. 252 Chapter 15 – Bibliography Riediger, T., H. A. Schmid, et al. (2002). "Amylin and glucose co-activate area postrema neurons of the rat." Neurosci Lett 328(2): 121-4. Riediger, T., H. A. Schmid, et al. (1999). "Pharmacological characterisation of amylin-related peptides activating subfornical organ neurones." Brain Res 837(1-2): 161-8. Riezzo, G., F. Indrio, et al. (2000). "Gastric electrical activity and gastric emptying in term and preterm newborns." Neurogastroenterol Motil 12(3): 2239. Riezzo, G., F. Indrio, et al. (2009). "Maturation of gastric electrical activity, gastric emptying and intestinal permeability in preterm newborns during the first month of life." Ital J Pediatr 35(1): 6. Rink, T. J., K. Beaumont, et al. (1993). "Structure and biology of amylin." Trends Pharmacol Sci 14(4): 113-8. Rizzo, T. A., B. E. Metzger, et al. (1997). "Early malnutrition and child neurobehavioral development: insights from the study of children of diabetic mothers." Child Dev 68(1): 26-38. Rizzo, T. A., B. L. Silverman, et al. (1997). "Behavioral adjustment in children of diabetic mothers." Acta Paediatr 86(9): 969-74. Roberts, A. N., B. Leighton, et al. (1989). "Molecular and functional characterization of amylin, a peptide associated with type 2 diabetes mellitus." Proc Natl Acad Sci U S A 86(24): 9662-6. Rouge, C., H. Piloquet, et al. (2009). "Oral supplementation with probiotics in very-low-birth-weight preterm infants: a randomized, double-blind, placebocontrolled trial." Am J Clin Nutr 89(6): 1828-35. Rowan, J. A., W. Gao, et al. (2010). "Glycemia and its relationship to outcomes in the metformin in gestational diabetes trial." Diabetes Care 33(1): 9-16. 253 Chapter 15 – Bibliography Rowan, J. A., W. M. Hague, et al. (2008). "Metformin versus insulin for the treatment of gestational diabetes." N Engl J Med 358(19): 2003-15. Rowland, N. E. and R. M. Richmond (1999). "Area postrema and the anorectic actions of dexfenfluramine and amylin." Brain Res 820(1-2): 86-91. Ryan, G. J., L. J. Jobe, et al. (2005). "Pramlintide in the treatment of type 1 and type 2 diabetes mellitus." Clin Ther 27(10): 1500-12. Ryle, A. P., F. Sanger, et al. (1955). "The disulphide bonds of insulin." Biochem J 60(4): 541-56. Sabata, V., H. Frerichs, et al. (1970). "Insulin and glucose levels in umbilical cord blood after infusions of glucose and glucose with insulin to women in labour." J Obstet Gynaecol Br Commonw 77(2): 121-8. Salhotra, A. and S. Ramji (2004). "Slow versus fast enteral feed advancement in very low birth weight infants: a randomized control trial." Indian Pediatr 41(5): 435-41. Samsom, M., L. M. Akkermans, et al. (1997). "Gastrointestinal motor mechanisms in hyperglycaemia induced delayed gastric emptying in type I diabetes mellitus." Gut 40(5): 641-6. Samsom, M., J. M. Roelofs, et al. (1998). "Proximal gastric motor activity in response to a liquid meal in type I diabetes mellitus with autonomic neuropathy." Dig Dis Sci 43(3): 491-6. Samsom, M., L. A. Szarka, et al. (2000). "Pramlintide, an amylin analog, selectively delays gastric emptying: potential role of vagal inhibition." Am J Physiol Gastrointest Liver Physiol 278(6): G946-51. Samsom, M., J. R. Vermeijden, et al. (2003). "Prevalence of delayed gastric emptying in diabetic patients and relationship to dyspeptic symptoms: a prospective study in unselected diabetic patients." Diabetes Care 26(11): 311622. 254 Chapter 15 – Bibliography Sanaka, M., Y. Koike, et al. (1997). "A reliable and convenient parameter of the rate of paracetamol absorption to measure gastric emptying rate of liquids." Int J Clin Pharmacol Ther 35(11): 509-13. Sanke, T., T. Hanabusa, et al. (1991). "Plasma islet amyloid polypeptide (Amylin) levels and their responses to oral glucose in type 2 (non-insulindependent) diabetic patients." Diabetologia 34(2): 129-32. Sari, F. N., E. A. Dizdar, et al. (2011). "Oral probiotics: Lactobacillus sporogenes for prevention of necrotizing enterocolitis in very low-birth weight infants: a randomized, controlled trial." Eur J Clin Nutr 65(4): 434-9. Sase, M., I. Miwa, et al. (2005). "Gastric emptying cycles in the human fetus." Am J Obstet Gynecol 193(3 Pt 2): 1000-4. Satoskar, A. A., K. Burdge, et al. (2007). "Typing of amyloidosis in renal biopsies: diagnostic pitfalls." Arch Pathol Lab Med 131(6): 917-22. Savino, F., F. Cresi, et al. (2004). "Gastric emptying in infants: epigastric impedance versus scintigraphy." Acta Paediatr 93(5): 608-12. Scammon RE, D. L. (1920). "Observations on the capacity of the stomach in the first ten days of postnatal life." Am J Dis Child 20: 516-538. Schanler, R. J., R. J. Shulman, et al. (1999). "Feeding strategies for premature infants: randomized trial of gastrointestinal priming and tube-feeding method." Pediatrics 103(2): 434-9. Schirra, J., M. Katschinski, et al. (1996). "Gastric emptying and release of incretin hormones after glucose ingestion in humans." J Clin Invest 97(1): 92103. Schirra, J., P. Leicht, et al. (1998). "Mechanisms of the antidiabetic action of subcutaneous glucagon-like peptide-1(7-36)amide in non-insulin dependent diabetes mellitus." J Endocrinol 156(1): 177-86. 255 Chapter 15 – Bibliography Schmid, H. A., M. Rauch, et al. (1998). "Effect of calcitonin on the activity of ANG II-responsive neurons in the rat subfornical organ." Am J Physiol 274(6 Pt 2): R1646-52. Schmitz, O., B. Brock, et al. (2004). "Amylin agonists: a novel approach in the treatment of diabetes." Diabetes 53 Suppl 3: S233-8. Schvarcz, E., M. Palmer, et al. (1997). "Physiological hyperglycemia slows gastric emptying in normal subjects and patients with insulin-dependent diabetes mellitus." Gastroenterology 113(1): 60-6. Schvarcz, E., M. Palmer, et al. (1993). "Hypoglycaemia increases the gastric emptying rate in patients with type 1 diabetes mellitus." Diabet Med 10(7): 6603. Schwartz, J. G., G. M. Green, et al. (1996). "Rapid gastric emptying of a solid pancake meal in type II diabetic patients." Diabetes Care 19(5): 468-71. Schwizer, W., H. Maecke, et al. (1992). "Measurement of gastric emptying by magnetic resonance imaging in humans." Gastroenterology 103(2): 369-76. Sedman, V. L., S. Allen, et al. (2005). "Atomic force microscopy study of human amylin (20-29) fibrils." Protein Pept Lett 12(1): 79-83. Sexton, P. M., G. Paxinos, et al. (1994). "In vitro autoradiographic localization of amylin binding sites in rat brain." Neuroscience 62(2): 553-67. Shapiro, R. E. and R. R. Miselis (1985). "The central neural connections of the area postrema of the rat." J Comp Neurol 234(3): 344-64. Sheskin, D. J. (2003). Handbook of parametric and nonparametric statistical procedures, Boca Raton: Chapman & Hall /CRC. Shulman, R. J. (2002). "Effect of enteral administration of insulin on intestinal development and feeding tolerance in preterm infants: a pilot study." Arch Dis Child Fetal Neonatal Ed 86(2): F131-3. 256 Chapter 15 – Bibliography Sidebottom, R., J. S. Curran, et al. (1983). "Effects of long-chain vs mediumchain triglycerides on gastric emptying time in premature infants." J Pediatr 102(3): 448-50. Sidery, M. B., I. A. Macdonald, et al. (1994). "Superior mesenteric artery blood flow and gastric emptying in humans and the differential effects of high fat and high carbohydrate meals." Gut 35(2): 186-90. Siegel, M., B. Krantz, et al. (1985). "Effect of fat and carbohydrate composition on the gastric emptying of isocaloric feedings in premature infants." Gastroenterology 89(4): 785-90. Siegel, M., E. Lebenthal, et al. (1984). "Effect of caloric density on gastric emptying in premature infants." J Pediatr 104(1): 118-22. Siegel, M., E. Lebenthal, et al. (1982). "Gastric emptying in prematures of isocaloric feedings with differing osmolalities." Pediatr Res 16(2): 141-7. Silverman, B. L., B. E. Metzger, et al. (1995). "Impaired glucose tolerance in adolescent offspring of diabetic mothers. Relationship to fetal hyperinsulinism." Diabetes Care 18(5): 611-7. Silvestre, M. A., C. A. Morbach, et al. (1996). "A prospective randomized trial comparing continuous versus intermittent feeding methods in very low birth weight neonates." J Pediatr 128(6): 748-52. Silvestre, R. A., J. Rodriguez-Gallardo, et al. (2001). "Selective amylin inhibition of the glucagon response to arginine is extrinsic to the pancreas." Am J Physiol Endocrinol Metab 280(3): E443-9. Slagle, T. A. and S. J. Gross (1988). "Effect of early low-volume enteral substrate on subsequent feeding tolerance in very low birth weight infants." J Pediatr 113(3): 526-31. 257 Chapter 15 – Bibliography Stanley, K., A. Magides, et al. (1995). "Delayed gastric emptying as a factor in delayed postprandial glycaemic response in pregnancy." Br J Obstet Gynaecol 102(4): 288-91. Sumner Makin, O. and L. C. Serpell (2004). "Structural characterisation of islet amyloid polypeptide fibrils." J Mol Biol 335(5): 1279-88. Symlin, p. (2005). Symlin [package insert]. San Diego, CA: Amylin Pharmaceuticals; June 2005. Szarka, L. A., M. Camilleri, et al. (2008). "A stable isotope breath test with a standard meal for abnormal gastric emptying of solids in the clinic and in research." Clin Gastroenterol Hepatol 6(6): 635-643 e1. Tan-Dy, C. R. and A. Ohlsson (2005). "Lactase treated feeds to promote growth and feeding tolerance in preterm infants." Cochrane Database Syst Rev(2): CD004591. Thomas, K., M. de Gasparo, et al. (1967). "Insulin levels in the umbilical vein and in the umbilical artery of newborns of normal and gestational diabetic mothers." Diabetologia 3(3): 299-304. Thomaseth, K., A. Kautzky-Willer, et al. (1996). "Integrated mathematical model to assess beta-cell activity during the oral glucose test." Am J Physiol 270(3 Pt 1): E522-31. Tomomasa, T., Z. Itoh, et al. (1985). "Nonmigrating rhythmic activity in the stomach and duodenum of neonates." Biol Neonate 48(1): 1-9. Torsdottir, I., M. Alpsten, et al. (1991). "A small dose of soluble alginate-fiber affects postprandial glycemia and gastric emptying in humans with diabetes." J Nutr 121(6): 795-9. 258 Chapter 15 – Bibliography Toshimori, H., R. Narita, et al. (1990). "Islet amyloid polypeptide (IAPP) in the gastrointestinal tract and pancreas of man and rat." Cell Tissue Res 262(3): 401-6. Tougas, G., Y. Chen, et al. (2000). "Standardization of a simplified scintigraphic methodology for the assessment of gastric emptying in a multicenter setting." Am J Gastroenterol 95(1): 78-86. Tschop, M., D. L. Smiley, et al. (2000). "Ghrelin induces adiposity in rodents." Nature 407(6806): 908-13. van der Kooy, D. and L. Y. Koda (1983). "Organization of the projections of a circumventricular organ: the area postrema in the rat." J Comp Neurol 219(3): 328-38. van Rossum, D., D. P. Menard, et al. (1994). "Autoradiographic distribution and receptor binding profile of [125I]Bolton Hunter-rat amylin binding sites in the rat brain." J Pharmacol Exp Ther 270(2): 779-87. van Wyk, M., D. K. Sommers, et al. (1995). "The proportional cumulative area under the curve of paracetamol used as an index of gastric emptying in diabetic patients with symptoms of gastroparesis." Clin Exp Pharmacol Physiol 22(9): 637-40. Vela-Huerta, M. M., E. U. San Vicente-Santoscoy, et al. (2008). "Leptin, insulin, and glucose serum levels in large-for-gestational-age infants of diabetic and non-diabetic mothers." J Pediatr Endocrinol Metab 21(1): 17-22. Vella, A., J. S. Lee, et al. (2002). "Effects of pramlintide, an amylin analogue, on gastric emptying in type 1 and 2 diabetes mellitus." Neurogastroenterol Motil 14(2): 123-31. Victor, Y. H. (1975). "Effect of body position on gastric emptying in the neonate." Arch Dis Child 50(7): 500-4. 259 Chapter 15 – Bibliography Villa, I., A. Rubinacci, et al. (1997). "Effects of amylin on human osteoblast-like cells." Peptides 18(4): 537-40. Wang, L. Y., H. Y. Hung, et al. (1997). "Clinical experience with early enteral feeding in very-low-birth-weight infants." Zhonghua Min Guo Xiao Er Ke Yi Xue Hui Za Zhi 38(4): 282-7. Westerbeek, E. A., R. L. Hensgens, et al. (2011). "The effect of neutral and acidic oligosaccharides on stool viscosity, stool frequency and stool pH in preterm infants." Acta Paediatr 100(11): 1426-31. Westermark, P. (1973). "Fine structure of islets of Langerhans in insular amyloidosis." Virchows Arch A Pathol Pathol Anat 359(1): 1-18. Westermark, P., C. Wernstedt, et al. (1987). "Islet amyloid in type 2 human diabetes mellitus and adult diabetic cats contains a novel putative polypeptide hormone." Am J Pathol 127(3): 414-7. Weyer, C., D. G. Maggs, et al. (2001). "Amylin replacement with pramlintide as an adjunct to insulin therapy in type 1 and type 2 diabetes mellitus: a physiological approach toward improved metabolic control." Curr Pharm Des 7(14): 1353-73. Whitehouse, F., D. F. Kruger, et al. (2002). "A randomized study and open-label extension evaluating the long-term efficacy of pramlintide as an adjunct to insulin therapy in type 1 diabetes." Diabetes Care 25(4): 724-30. Wimalawansa, S. J. (1997). "Amylin, calcitonin gene-related peptide, calcitonin, and adrenomedullin: a peptide superfamily." Crit Rev Neurobiol 11(2-3): 167239. Woerle, H. J., M. Albrecht, et al. (2008). "Importance of changes in gastric emptying for postprandial plasma glucose fluxes in healthy humans." Am J Physiol Endocrinol Metab 294(1): E103-9. 260 Chapter 15 – Bibliography Wookey, P. J., Z. Cao, et al. (1998). "Interaction of the renal amylin and reninangiotensin systems in animal models of diabetes and hypertension." Miner Electrolyte Metab 24(6): 389-99. Wookey, P. J., Z. Cao, et al. (1997). "Increased density of renal amylin binding sites in experimental hypertension." Hypertension 30(3 Pt 1): 455-60. Wookey, P. J. and M. E. Cooper (1998). "Amylin: physiological roles in the kidney and a hypothesis for its role in hypertension." Clin Exp Pharmacol Physiol 25(9): 653-60. Wookey, P. J., C. Tikellis, et al. (1996). "Amylin binding in rat renal cortex, stimulation of adenylyl cyclase, and activation of plasma renin." Am J Physiol 270(2 Pt 2): F289-94. Wookey, P. J., C. Tikellis, et al. (1998). "Amylin as a growth factor during fetal and postnatal development of the rat kidney." Kidney Int 53(1): 25-30. Wren, A. M., L. J. Seal, et al. (2001). "Ghrelin enhances appetite and increases food intake in humans." J Clin Endocrinol Metab 86(12): 5992. Wren, A. M., C. J. Small, et al. (2001). "Ghrelin causes hyperphagia and obesity in rats." Diabetes 50(11): 2540-7. Yang, J., E. A. Cummings, et al. (2006). "Fetal and neonatal outcomes of diabetic pregnancies." Obstet Gynecol 108(3 Pt 1): 644-50. Young, A. (1997). "Role of amylin in nutrient intake - animal studies." Diabet Med 14 Suppl 2: S14-8. Young, A. (2005). "Effects on digestive secretions." Adv Pharmacol 52: 123-50. Young, A. (2005). "Inhibition of gastric emptying." Adv Pharmacol 52: 99-121. Young, A. (2005). "Inhibition of glucagon secretion." Adv Pharmacol 52: 151-71. 261 Chapter 15 – Bibliography Young, A. (2005). "Receptor pharmacology." Adv Pharmacol 52: 47-65. Young, A., R. Pittner, et al. (1995). "Amylin regulation of carbohydrate metabolism." Biochem Soc Trans 23(2): 325-31. Young, A. A., B. Gedulin, et al. (1994). "Selective amylin antagonist suppresses rise in plasma lactate after intravenous glucose in the rat. Evidence for a metabolic role of endogenous amylin." FEBS Lett 343(3): 237-41. Young, A. A., B. Gedulin, et al. (1995). "Gastric emptying is accelerated in diabetic BB rats and is slowed by subcutaneous injections of amylin." Diabetologia 38(6): 642-8. Young, A. A., B. R. Gedulin, et al. (1996). "Dose-responses for the slowing of gastric emptying in a rodent model by glucagon-like peptide (7-36) NH2, amylin, cholecystokinin, and other possible regulators of nutrient uptake." Metabolism 45(1): 1-3. Young, A. A., T. J. Rink, et al. (1993). "Dose response characteristics for the hyperglycemic, hyperlactemic, hypotensive and hypocalcemic actions of amylin and calcitonin gene-related peptide-I (CGRP alpha) in the fasted, anaesthetized rat." Life Sci 52(21): 1717-26. Zhu, G. C., D. T. Dudley, et al. (1991). "Amylin increases cyclic AMP formation in L6 myocytes through calcitonin gene-related peptide receptors." Biochem Biophys Res Commun 177(2): 771-6. Ziegler, D., P. Schadewaldt, et al. (1996). "[13C]octanoic acid breath test for non-invasive assessment of gastric emptying in diabetic patients: validation and relationship to gastric symptoms and cardiovascular autonomic function." Diabetologia 39(7): 823-30. 262