ASH NEWS AND REPORTS® May/june 2014 Volume 11 Issue 3 Newly Redesigned ASH Website Offers Improved Search Capability and Access to Most-Viewed Content If you have visited the ASH website in the past few weeks you may have noticed some significant changes. A new site design was launched on April 4 as part of a multiyear effort to upgrade the Society’s technology platforms. ASH President Linda J. Burns, MD, explained, “The ASH leadership felt the time had come to invest in a strategy that will better serve the Society’s membership by bringing ASH’s digital offerings in closer alignment with member needs.” These offerings include not only the Society’s website, but also mobile apps, e-books, and other forms of electronic communication. The first step in the process entailed months of research to better understand why ASH’s key audiences visit the website, the specific tasks they are trying to accomplish, and how successful they are. “By using software to track where users tend to click most often throughout ASH’s website, we learned that many of the navigational headings designed to help users find information quickly were not being utilized,” said Michael Mersky, ASH’s Web Manager. “So we employed a number of user testing exercises to find out what terms our users would choose to name and (Cont. on page 6) D I F F U S I O N Features A Hereditary Bleeding Disorder Reveals a New Cog in the Coagulation Mechanism 4 Vincent LM, Tran S, Livaja R, et al. Coagulation factor VA2440G causes east Texas bleeding disorder via TFPIα. J Clin Invest. 2013;123:3777-3787. Ask the Hematologist – Dr. Janet Abrahm discusses her approach to the management of chemotherapy-induced peripheral neuropathy. 7 Update on the 2013 ASH Choosing Wisely Initiative – Dr. Lisa Hicks reviews the ASH Choosing Wisely initiative and summarizes the ongoing aims of the ASH Choosing Wisely Task Force. 14 My Journey in Hematology – Dr. John Adamson recounts highlights of his remarkable career that has taken him on a circuitous journey from California to Oxford and back again. DEPARTMENTS 2president’s column 3News and Reports 7The Hematologist Advocate W hat is the differential diagnosis in a 35-year-old male with a lifelong history of easy bruising, epistaxis, and prolongation of both the prothrombin time (PT) and the activated partial thromboplastin time (APTT)? So began the evaluation by Dr. Shao-Qing Kuang et al. in the laboratory of Dr. Dianna Milewicz at the University of Texas Health Science Center in Houston, Texas.1 The family history revealed an autosomal bleeding disorder in an east Texas kindred consisting of 46 family members spanning four generations. Surprisingly, coagulation factor assays for fibrinogen and factors II, V, VII, VIII, IX, X, XI, and XII were normal. Linkage analysis demonstrated that the factor V (FV) gene (F5) mapped to the disease interval. Sequencing of the F5 gene revealed an A2440G mutation in exon 13 at position 2440, predicting a S756G mutation in the B domain of FV. However, because FV levels were normal, the mutation was considered an unlikely cause of the bleeding diathesis. The cause of this mysterious bleeding disorder remained unknown for more than another decade until it was elucidated by Dr. Lisa Vincent and colleagues from the laboratories of Dr. Milewicz in Houston and Dr. Björn Dahlbäck of Lund University Hospital in Malmö, Sweden. The authors began by looking for abnormalities in the plasma FV in the kindred by Western blotting. Full-length FV is 330 kDa procofactor that is proteolytically activated by thrombin or factor Xa. During this process, the large, central B domain is removed (Figure). The product, FVa, is a heavy chain/light chain heterodimer that is a cofactor for Figure 8Diffusion 13Clinical Trials Corner Full-length FV HEAVY C HAIN LIGHT C HAIN 15EDITORS’ CHOICE B 16WHAT’S ON THE WEB BR FV-short AR HEAVY C HAIN LIGHT C HAIN B AR Full-length FV is a 330 kDa protein that contains a large central B domain. An acidic region (AR) and a basic region (BR) are involved in the control of the activation of FV. FV-short is produced by alternative splicing of the FV mRNA, leading to deletion of ~85% of the B domain. TFPIα binds FV-short with higher affinity than full-length FV. Increased levels of FV-short due to the A2440G mutation in the F5 gene of the east Texas kindred results in increased plasma levels of TFPIα and a bleeding diathesis. peter lollar, MD Dr. Lollar indicated no relevant conflicts of interest. factor Xa in the prothrombinase complex. Analysis of 36 family members revealed that in addition to full-length FV, plasma from affected family members contained a prominent ~250 kDa variant, which they named FV-short. FV-short was also identified in much smaller amounts in unaffected family members. RT-PCR analysis of whole blood from affected family members using primers spanning exon 13 identified an expected 2946 bp product and, additionally, large amounts of a smaller 840 bp product. The 840 bp fragment was also found in unaffected family members but, as with FV-short protein, in much smaller amounts. Sequencing of the 840 bp product revealed that the F5 gene variant produces an in-frame deletion of 2,106 base pairs, resulting in the deletion of nearly 85 percent of the B domain (Figure). A thrombin generation assay was used as a potentially more sensitive method to detect abnormalities in the tissue factor-initiated extrinsic coagulation pathway in the kindred. Despite having normal coagulation factor levels, all affected family members demonstrated decreased thrombin generation. The addition of recombinant FVshort to immunodepleted FV-deficient plasma resulted in normal thrombin generation, indicating that the abnormality was not due to an intrinsic procoagulant defect in FV-short per se. However, addition of affected plasma to unaffected plasma resulted in decreased thrombin generation, revealing the presence of a coagulation inhibitor in affected plasma. To identify the inhibitor, the authors took advantage of the recent discovery by Dr. Connie Duckers et al. that FV binds to tissue factor pathway inhibitor α (TFPIα).2 This finding was a result of the clinical observation that patients with severe FV deficiency often only have a mild bleeding disorder. This finding suggests that FV deficiency produces a secondary deficiency of a coagulation inhibitor with which it is associated. Dr. Duckers et al. made the key observation that plasma levels of TFPIα, a candidate coagulation inhibitor, is decreased in FV deficient individuals. They then demonstrated that TFPIα binds FV in a purified system. These observations led Dr. Vincent et al. to the hypothesis that the inhibitor in affected family members is TFPIα and that the greater inhibitor activity must be a consequence of the high concentration of FV-short. The addition of anti-TFPIα antibodies to affected plasma increased thrombin generation to normal levels, indicating that TFPIα is the culprit producing the coagulant abnormality in the kindred. Furthermore, addition of anti-TFPIα antibodies to affected plasma removed FV-short, but not full-length FV, suggesting a stronger association of TFPIα with FV-short. This interpretation was confirmed by a semi-quantitative immunoprecipitation assay showing that TFPIα binds with higher affinity to FV-short than to full-length FV. The authors also found that TFPIα levels are increased several-fold in affected (Cont. on page 2) President’s Column Hematologist THE AS H N e w s a n d R e p o rt s ® I SSN 1 5 5 1 - 8 7 7 9 Training the Next Generation Editor-in-Chief: A Charles Parker, MD University of Utah Salt Lake City, UT I’m proud of the Society’s long history of partnering with medical educators in hematology to advance scholarship in our subspecialty by tackling head-on the challenges we’ve all had to face. From practical approaches such as training guidelines, to advocating for evidence-based medical education, to creation of tools for educators, to awarding of grants to trainees, ASH has stood by those of us on the front lines of our medical schools and teaching hospitals. Several new initiatives aim to continue this tradition: Pamela S. Becker, MD, PhD University of Washington Seattle, WA s a fellowship program director for the past 18 years, I marvel at the changes that have been introduced into the training environment. From managing mandated reductions in duty hours of fellows to documentation of competency measures, it seems those of us in the educational space have not had a moment’s rest! 1. As has been reported in this publication, in ASH NewsLink, and on the ASH website, the Committee on Training, chaired by Gary Schiller, MD, is interested in developing an ASH Medical Educators Institute. This program, modeled on ASH’s Clinical Research Training Institute, will provide guidance to dedicated hematology educators who are aiming both to improve individual teaching skills and to enhance learning opportunities at their home institution. The committee and I would be interested in your thoughts on crafting this program and count on your enthusiastic volunteerism in making this new initiative a success. 2. A working group, organized by the Committee on Training and led by Elaine Muchmore, MD, is in the process of developing milestones for fellows in hematology training programs. Creation of these milestones is a critical element necessary to meet the requirements of the Accreditation Council for Graduate Medical Education’s Next Accreditation System (ACGME NAS). For those of you who have yet to hear of this sweeping new program, NAS began phase implementation in July 2013 and represents the culmination of a multi-year process of restructuring ACGME’s accreditation system based on educational outcomes in clinical competency. 3. The Teaching Cases Subcommittee, which has responsibility for developing and maintaining the ever-popular medical student cases on the ASH website, has been diligently developing new cases and researching new approaches to make the cases more interactive and engaging. This program is strongly supported by the Society as evidence by its prominent place on the agenda of the upcoming ASH Executive Committee meeting. 4. Last spring, the Society launched the Fundamentals for Hematology Fellows Program that provides a bundle of ASH resources to fellows in hematology-related training programs. This initiative has exceeded our expectations by adding more than 1,000 additional fellows to our Associate member category. The program not only provides a connection from ASH to the trainee, but also makes participants eligible for ASH programs such as the Clinical Research Training Institute. These new efforts join a panoply of continuing programs, including the Research Training Award for Fellows, annual meeting events such as the Hematology Course Directors’ Workshop, and services such as the Hematology In-Service Examination, designed to address hematology training from the perspective of the learner, the educator, and the training environment. I hope every educator in hematology knows about these programs and uses them to their best advantage. Please contact training@hematology.org with any questions about ASH’s educational programs. Our goal at ASH is to enthusiastically support medical educators in hematology and, together with this dedicated group, to continue improving training in our subspecialty by developing new initiatives and sustaining ongoing programs that focus on the needs of faculty and fellows. Contributing Editors: Theresa Coetzer, PhD University of the Witwatersrand Johannesburg, South Africa Adam Cuker, MD, MS University of Pennsylvania Philadelphia, PA David Garcia, MD University of Washington Seattle, WA Jason Gotlib, MD, MS Stanford Cancer Center Stanford, CA Peter Johnson, MD Southampton General Hospital Southampton, United Kingdom Mark J. Koury, MD Vanderbilt University Nashville, TN Peter Kurre, MD Oregon Health & Science University Portland, OR Ann LaCasce, MD, MSc Dana-Farber Cancer Institute Boston, MA Pete Lollar, MD Emory Children’s Center Atlanta, GA Charles T. Quinn, MD, MS Cincinnati Children’s Hospital Medical Center Cincinnati, OH Gregory M. Vercellotti, MD University of Minnesota Minneapolis, MN Linda J. Burns, MD Managing Editor Karen Learner Editorial Assistant Tiffany Reid, MA Graphic Designer A Hereditary Bleeding Disorder Reveals a New Cog in the Coagulation Mechanism D I F F U S I O N The authors speculated that the normal levels of FV and other coagulation factors in affected plasmas are due to the strong procoagulant stimulus in coagulation assays (tissue factor in the PT and activated partial thromboplastin in the APTT). Accordingly, the rapid production of high concentrations of Factor VIIa and Factor Xa would overwhelm TFPIα, even when its concentration is elevated in affected plasmas. In contrast, thrombin generation assays were performed in diluted plasma and at low concentrations of tissue factor, leading to more sensitive detection of a coagulant defect due to elevated levels of TFPIα. The role of the large B domain in FV function has been enigmatic. Recently, Dr. Mettine H.A. Bos and Dr. Rodney M. Camire demonstrated that the B domain serves to maintain FV in an inactive, procofactor state.3 A basic region and an acidic region in the B domain have been identified that are necessary for this function (Figure). Interestingly, the basic region is not present in FV-short (Figure), which has implications for FV and TFPIα structure and function that remain to be investigated. 2 American Society of Hematology 2021 L Street, NW, Suite 900 Washington, DC 20036 klearner@hematology.org (Cont. from page 1) plasmas compared with unaffected plasmas. They proposed that free, 40 kDa TFPIα is cleared by kidneys, and that this clearance is prevented when TFPIα is bound in a high-molecular-weight complex with FV or FV-short. Thus, the increased association of TFPIα with FVshort leads to higher plasma levels. grayHouse design An A2440G mutation in exon 13 of the F5 gene results in increased usage of an alternative splice-donor site, leading to production of a previously undetected protein, FV-short. Alternative splicing and expression of FV-short also occurs in the absence of the mutation, albeit to a lesser extent. TFPIα binds to full-length FV and FV-short, but with higher affinity to the latter. The association of TFPIα with FV-short reduces the clearance of TFPIα by preventing renal filtration. The resulting increase in TFPIα produces a bleeding diathesis due to an increased inhibition of Factor VIIa and Factor Xa. The results of this study have identified an important new mechanisms underlying FV and TFPIα biology in the regulation of coagulation. 1. Kuang S, Hasham S, Phillips MD, et al. Characterization of a novel autosomal dominant bleeding disorder in a large kindred from east Texas. Blood. 2001;97:1549-1554. 2. Duckers C, Simioni P, Spiezia L, et al. Low plasma levels of tissue factor pathway inhibitor in patients with congenital factor V deficiency. Blood. 2008;112:3615-3623. ©2014 by the American Society of Hematology. All materials contained in this newsletter are protected by copyright laws and may not be used, reproduced, or otherwise exploited in any manner without the express prior written permission of The Hematologist: ASH News and Reports. Any third-party materials communicated to The Hematologist become its copyrighted property and may be used, reproduced, or otherwise exploited by The Hematologist. Contributing authors have declared any financial interest in a product or in potentially competing products, regardless of the dollar amount. Any such financial interest is noted at the bottom of the article. Dr. Parker has no relevant conflicts of interest to disclose. 3. Bos MH and Camire RM. A bipartite autoinhibitory region within the B-domain suppresses function in factor V. J Biol Chem. 2012;287:26342-26351. The Hematologist: ASH News and Reports n e ws and r e p or t s Register to Attend the New ASH Meeting on Lymphoma Biology August 10-13, 2014 The Broadmoor, Colorado Springs, Colorado Get a comprehensive update on the most recent discoveries in lymphoma biology and interact with world-class experts in the field. During this three-day forum, keynote speaker Klaus Rajewsky, MD, will discuss immune regulation and cancer, and attendees will hear about the most recent progress in basic and translational lymphoma research, share their findings with colleagues, and exchange ideas with experts on how to move the field forward. Klaus Rajewsky, MD What to Expect • Informal setting with networking opportunities to share ideas and establish collaborations • Presentations by invited experts on topics including: • B-cell receptor signaling • Germinal center biology • Epigenetics of lymphoma • Immune evasion • Lymphoma genetics • Metabolism • T-cell lymphoma • Panel discussions • Oral presentations of the top-rated abstracts • Poster sessions and interactive workshops For more information about the meeting and to register, go to www.hematology. org/Lymphoma-Biology. 2014 ASH Annual Meeting and Exposition December 6-9 Moscone Center, San Francisco, CA The American Society of Hematology (ASH) invites you to save the date for the 56th ASH Annual Meeting in San Francisco, CA. As the premier hematology event, this meeting will provide attendees with an invaluable educational experience and the opportunity to: • Review more than 3,000 scientific abstracts. • Interact with the global community of more than 20,000 hematology professionals from every subspecialty. • A ttend the Education and Scientific Program sessions that feature lectures by leaders in the field. • D evelop collaborations with clinical and basic investigators who share your research interests. • S hare the experience with friends and colleagues in a collegial, stimulating atmosphere in one of America’s great cities. Key Dates Abstract submission site opensJune 5 Advance member registration & housingJuly 24 ASH and ASCO Present New Seminar on Hematology for the Oncologist ASH is partnering with the American Society of Clinical Oncology (ASCO) to present a Hematology for the Oncologist symposium, May 2930, 2014, in Chicago, IL, as part of ASCO’s Pre-Annual Meeting Seminars. Lectures will focus on hematologic issues that medical oncologists commonly encounter in a consultative practice. Experts in the field will discuss clinical aspects of bleeding and clotting disorders, novel oral anticoagulants, iron replacement therapy, and other benign hematology topics. Question-and-answer periods will be incorporated into the sessions to encourage discussion and ensure maximum interaction between faculty and participants. To learn more about the symposium, go to http://am.asco.org/ hematology-oncologist-seminar. Advance registration for non-membersAugust 13 The Hematologist: ASH News and Reports 3 ASH does not recommend or endorse any specific tests, physicians, products, procedures, or opinions, and disclaims any representation, warranty, or guaranty as to the same. Reliance on any information provided in this article is solely at your own risk. Ask the Hematologist janet l. abrahm, md Professor of Medicine, Harvard Medical School; Division of Adult Palliative Care, Department of Psychosocial Oncology and Palliative Care, Dana-Farber Cancer Institute The Case The patient is a 54-year-old woman with IgA lambda multiple myeloma, receiving dexamethasone, lenalidomide, and bortezomib therapy. She was healthy before her diagnosis but had mild hypercalcemia and mild renal insufficiency at diagnosis, both of which have resolved with treatment. She is about to begin her third treatment cycle, but reports painful (pain score of 7/10) numbness and paresthesias in her fingers and feet. The former problem causes clumsiness when using her touch-screen tablet and phone because she cannot accurately feel her fingers touch the screens, and she needs the light on at night because she cannot reliably feel when her feet touch the floor. Her Zubrod (ECOG) performance status is 1. She had similar, but less severe, symptoms at the beginning of her last chemotherapy cycle, but she did not mention them to anyone for fear that treatment would be delayed or discontinued. NCI-CTC for Neuropathy Peripheral Sensory Neuropathy Grade 1: Asymptomatic or loss of deep tendon reflexes or paresthesia Grade 2: Moderate symptoms limiting instrumental activities of daily living (ADL) Grade 3: Severe symptoms limiting self-care ADL Grade 4: Life-threatening consequences; urgent intervention indicated Grade 5: Death Peripheral Motor Neuropathy Grade 1: Asymptomatic clinical or diagnostic observations only; intervention not indicated Grade 2: Moderate symptoms limiting instrumental ADL Grade 3: Severe symptoms limiting self-care ADL; assistive device indicated During her exam, she was anxious, alert, and oriented. She denied both spine pain and bowel or bladder incontinence. Her exam was unremarkable except for the neurologic examination. She had impaired proprioception, decreased vibration and light touch in her hands to the wrists and feet to the ankles, 4/5 hand grip and finger strength bilaterally, and 4/5 ankle strength. Her ankle deep tendon reflexes (DTRs) were absent (flaccid). Laboratory studies showed normal potassium, BUN, creatinine, glucose, calcium, and magnesium. B12 concentration and thyroid function studies were normal. Her paraprotein concentration showed a continuing decrease in response to treatment. The Question What is your approach to assessment and management of patients with chemotherapyinduced peripheral neuropathy? My Response Presentation/Physical Findings The patient’s presentation is typical of chemotherapy-induced peripheral neuropathy (CIPN) that is characterized by tingling (71%), numbness (58%), and paresthesias or dysesthesias (45%), or spontaneous burning, cramping, or aching pain (40%) that is usually symmetric, begins peripherally and progresses proximally.1 Patients often report allodynia (i.e., pain from something that is usually not painful, like cold temperatures or the touch of clothing), or hyperalgesia (i.e., experiencing more pain than usual from a mildly painful stimulus). The mechanisms of axonal nerve injury vary by agent and include damage to mitochondria (bortezomib, platinum compounds, vincristine, paclitaxel), glial cells and macrophages (bortezomib, oxaliplatin, vincristine, paclitaxel), vasa vasorum (thalidomide, paclitaxel), endoplasmic reticulum (bortezomib), or microtubules (bortezomib, taxanes, and vinca alkaloids).2,3 CIPN induced by vinca alkaloids causes numbness, not pain, and loss of DTRs, proprioception, and motor impairment, such as foot drop.2 Patients receiving thalidomide, bortezomib, or platinum have a mainly sensory neuropathy, which may include loss of vibration, proprioception, and DTRs.2 Taxanes cause both a sensory neuropathy and proximal weakness.2 Only CIPN induced by thalidomide is irreversible. Evaluation In a patient whose CIPN appears only after chemotherapy, the evaluation should include laboratory testing for B12 deficiency, hypothyroidism, hypocalcemia, hypomagnesemia, and hyperglycemia (Figure 1). Because patients with leptomeningeal carcinomatosis or spinal cord compression can present with these symptoms, an MRI of the spine with contrast is recommended for patients with back pain. If the test is negative, an MRI of the brachial or lumbosacral plexus, depending on the location of the symptoms and clinical suspicion, may be warranted. Diagnosis of CIPN is clinical, and nerve conduction studies, EMGs, and skin biopsies are not needed for diagnosis or management. Chemotherapy doses are generally adjusted for patients with grade 3 or greater neuropathy based on NCI common terminology criteria (CTC) for adverse events. 4 Grade 4: Life-threatening consequences; urgent intervention indicated Grade 5: Death The patient under discussion had grade 2 sensory neuropathy by these criteria, which underestimated her disability and functional losses. The NCI-CTC are not as sensitive to patient-reported symptoms or functional loss as are some other scoring systems, but no consensus has yet been reached on a standard for grading neurotoxicity.4,5 Quality-of-life scales may be more relevant (e.g., the CIPN 20 or FACT/GOG-Ntx).6 Treatment Dose reduction or drug discontinuation is the only specific therapy for most CIPNs; patients may tolerate higher doses of bortezomib if the drug is given by a subcutaneous injection rather than by IV infusion. Symptomatic therapies for CIPN are limited (Figure 1). Of the adjuvant agents effective for patients with neuropathic pain, only duloxetine (30-60 mg/day) has clearly shown efficacy for the CIPN induced by taxanes and platinum.7 Despite the lack of proven efficacy of other agents in CIPN, insurance companies usually require that patients first fail a trial of gabapentin or pregabalin before authorizing duloxetine. Occupational therapy or physical therapy is recommended for patients with ≥ grade 2 sensory or motor toxicity to improve function and quality of life. Patients should also be assessed for depression, which often accompanies uncontrolled chronic pain.8 While vitamin E, vitamin B6, magnesium, calcium, acetyl-L-carnitine, glutamine, glutathione, n-acetyl cysteine, omega-3 fatty acids, alpha lipoic acid, and cannabinoids each have shown efficacy in non-randomized, non-placebo-controlled trials, none can be recommended yet as standard therapy.1, 2, 9 Amitriptyline does not prevent or ameliorate chemotherapy-induced neuropathy from vinca alkaloids, platins, or taxanes.1 Venlafaxine, gabapentin, the N-methyl-d-aspartate (NMDA) receptor antagonist dextromethorphan, the oral anesthetics memantine and mexiletine, and low-dose continuous capsaicin treatment are also ineffective.2 Opioids are recommended for CIPN patients with severe pain.10 Patients will usually require a long-acting agent to provide basal pain relief (e.g., sustained-release morphine or a fentanyl patch) and short-acting opioids for breakthroughs. Methadone (Figure 2) is particularly helpful for basal pain relief. Methadone includes d- and l-isomers that act as opioid-receptor agonists and NMDA receptor antagonists.11 Because NMDA antagonizes the activity of the opiate receptors, blocking NMDA receptors enhances the analgesic effect of externally administered opioids. Methadone also inhibits the reuptake of serotonin and norepinephrine; therefore, it functions as a serotonin-norepinephrine reuptake inhibitor (SNRI). Methadone is usually given orally bid to tid. Dose adjustment is needed for hepatic failure but not for renal failure. The steady state is not reached until 72 hours after the initial dose or after a dose increase. Therefore, short-acting opioids should be used to control symptoms in the interim. If pain level falls to ≤ 3 within the first 24 to 48 hours of initiating methadone, reduce the dose by half immediately to avoid excessive sedation and respiratory depression that may otherwise occur in the next 24 hours. This delay in reaching steady-state plasma levels, along with potential cardiac toxicity (prolongation of the rate corrected QT [QTc] interval) and its interactions with inducers and inhibitors of the cytochrome P450 system, CYP1A2, CYP3A4, and CYP2D6 make using methadone safely somewhat complex. Palliative care specialists can be helpful in managing initiation or adjustment of methadone doses or for consulting on patients with seemingly refractory pain syndromes. The Hematologist: ASH News and Reports t h e p rac t icing h e ma t ologis t Evidence-Based Algorithm for Evaluation and Symptomatic Treatment of Patients with CIPN Figure 1 Figure 2 First, exclude spinal or leptomeningeal metastases and plexopathies and correct vitamin or hormone abnormalities, if present. For patients who have NCI-CTC grade ≥ 3 CIPN, follow standard protocol guidelines for dose adjustments of anti-neoplastic agents. Use duloxetine for patients with unacceptable pain levels who are receiving either platinum or taxanes. For refractory pain, add an opioid. If standard opioids are not effective, or if they cause excessive sedation, myoclonus, or other side effects, consider using methadone as the “basal” opioid, and use the standard opioids for breakthrough pain. *Despite the lack of proven efficacy, insurance companies may require that patients first fail a trial of gabapentin or prebabalin before authorizing duloxetine. Patients who, at baseline, have a prolonged QTc interval, or who need high doses of methadone, have developed cardiac arrhythmias (torsades de pointes; polymorphic ventricular tachycardia). Fatalities have been reported but were associated with doses of > 600 mg per day. Common drugs used in hematology patients that also prolong the QTc include the quinolone antibiotics (especially levofloxacin), typical and atypical antipsychotics (such as haloperidol and olanzapine), selective serotonin reuptake inhibitors (SSRIs) (but not SNRIs), and metoclopramide. Methadone’s drug interactions are listed on websites including www.drugs.com or www. qtdrug.org. Fluconazole, voriconazole, fluoxetine, and fluvoxamine raise methadone levels. SSRIs may raise methadone levels in CYP2D6 rapid metabolizers but SNRIs (e.g., venlafaxine) do not. Additionally, grapefruit juice and acute alcohol ingestion can increase methadone levels. Drug levels of desipramine (or other tricyclic antidepressants) and zidovudine increase when methadone is added. Antiretrovirals, phenobarbital, carbamazepine, phenytoin, rifampin, somatostatin, spironolactone, and risperidone lower the levels of methadone and have precipitated withdrawal symptoms. 1. Finnerup NB, Sindrup SH, Jensen TS. Management of painful neuropathies. Handb Clin Neurol. 2013;115:279-290. 2. Ferrier J, Pereira V, Busserolles J, et al. Emerging trends in understanding chemotherapy-induced peripheral neuropathy. Curr Pain Headache Rep. 2013;17:364-372. 3. Diezi M, Buclin T, Kuntzer T. Toxic and drug-induced peripheral neuropathies: updates on causes, mechanisms and management. Curr Opin Neurol 2013;26:481-488. 4.Alberti P, Rossi E, Cornblath DR, et al. Physician-assessed and patient-reported outcome measures in chemotherapy-induced sensory peripheral neurotoxicity: two sides of the same coin. Ann Oncol. 2014;25:257-264. 5. Driessen CML, de Kleine-Bolt KME, Vingerhoets AJJM, et al. Assessing the impact of chemotherapyinduced peripheral neurotoxicity on the quality of life of cancer patients. Support Care Cancer. 2012;20:877-881. 6. Grisold W, Cavaletti G, Windebank AJ. Peripheral neuropathies from chemotherapeutics and targeted agents: diagnosis, treatment, and prevention. Neuro Oncol. 2012;14:iv45-iv54. 7.Smith EML, Pang H, Cirrincione C, et al. Effect of duloxetine on pain, function, and quality of life among patients with chemotherapy-induced painful peripheral neuropathy: a randomized clinical trial. JAMA. 2013;309:1359-1367. 8. Glover J, Dibble SL, Dodd MJ, et al. Mood states of oncology outpatients: does pain make a difference? J Pain Symptom Manage. 1995;10:120-128. 9.Schloss JM, Colosimo M, Airey C, et al. Nutraceuticals and chemotherapy induced peripheral neuropathy (CIPN): a systematic review. Clin Nutr. 2013;32:888-893. 10. Dworkin RH, O’Connor AB, Audette J, et al. Recommendations for the pharmacologic management of neuropathic pain: an overview and literature update. Mayo Clin Proc. 2010;85:S3-S14. Conclusion The patient continued therapy, but bortezomib delivery was switched from IV infusion to SQ injection. Her pain symptoms fell to tolerable levels on 2 mg tid of methadone such that she was able to work, and her functional level was further enhanced by participation in an occupational therapy program. CIPN can be a disabling complication of chemotherapy. Patient education that encourages reporting relevant symptoms, chemotherapy dose adjustment, occupational therapy, physical therapy, and symptomatic therapy using duloxetine and opioids, including methadone, can often help patients maintain or regain function and minimize pain and disability. The Hematologist: To safely prescribe methadone, monitor the QTc. If the QTc is prolonged, correct contributing metabolic abnormalities or discontinue agents that can prolong the QTc. If the patient requires concomitant therapy with a CYP3A4 competitor (such as voriconazole), use lower doses of methadone. See text for other agents that require adjustments of methadone doses. Increase methadone doses no more than every 72 hours. ASH News and Reports 11. Rhondali W, Tremellat F, Ledoux M, et al. Methadone rotation for cancer patients with refractory pain in a palliative care unit: an observational study. J Pall Med 2013;16:1382-1387. Dr. Abrahm receives royalties from her book, A Physician’s Guide to Pain and Symptom Management in Cancer Patients, 2nd edition, 2005, from Johns Hopkins University Press. She is the paid section editor for UpToDate on the topic of pain in palliative care patients. She also receives expense reimbursement and honoraria from Knowledge to Practice for her lectures on palliative care. 5 Newly Redesigned ASH Website (Cont. from page 1) categorize various pieces of Web content, and we used those findings to develop a more intuitive navigational scheme.” meantime, please visit Hematology.org to explore the new design. We encourage you to share your feedback by emailing webmaster@hematology.org. Along with the upcoming Blood website redesign, more changes will be introduced in the coming months, as standalone sites such as the ASH Academy, an eLearning platform for hematologists, are brought into alignment with the look and feel of the new website. In addition, both the functionality of the ASH Store and the account management section of the ASH website will be enhanced throughout 2014. In the ASH staff will continue to engage in ongoing user testing to refine the website user experience. ASH members who are interested in being part of this process are encouraged to email webmaster@hematology.org to sign up for the Website Users Group. Members of this informal group will be invited to act as beta testers and to provide feedback prior to the rollout of new website features. Top 5 Website Changes You Should Know About: Website Organized Around Most-Accessed Resources – The site has been reorganized to showcase the content that is most popular with users. This content includes Blood, educational resources such as clinical practice guidelines, and information about ASH’s upcoming meetings, which are now front and center. Enhanced Viewing of The Hematologist Online – Readers will now be able to quickly get to their favorite sections and view collections of articles written by specific authors. ➔ ➔ Improved Search – Not only has ASH invested in a state-of-the-art search engine that provides far superior results compared with the previous search function, but users are now able to search across all ASH websites without leaving Hematology.org. In addition, users will see suggestions for related searches when the item they search for is not found. Responsive Design – The new website design adjusts automatically for optimal viewing from smartphones and tablet devices. Consistent Navigation – Later in 2014, all auxiliary ASH websites will have a consistent navigational structure for a more unified experience across sites. 6 The Hematologist: ASH News and Reports T H E H E M A T O L O G I S T A D V O C A TE HEADLINES FROM Washington Congress Moves Forward with FY 2015 Budget Process I n early March, President Obama kicked off the fiscal year (FY) budget and appropriations process with the release of his FY 2015 budget proposal. While noting that “biomedical research contributes to improving the health of the American people,” the President’s proposed budget seeks $30.2 billion for the National Institutes of Health (NIH), a slight increase over the amount provided for NIH in the final FY 2014 budget passed by Congress in January. The proposed budget requests small increases in funding for all of the Institutes of interest to hematology, including NHLBI, NCI, and NIDDK and estimates that this proposed increase in funding “would support about 650 additional new grants” NIH-wide. It is important to remember that the President’s nonbinding budget proposal merely sets forth the Administration’s priorities and is just one step in a lengthy federal budget process. Announced a month later than expected, and after the House and Senate had already established FY 2015 spending blueprints, the President’s budget begins a new round of congressional negotiations on annual spending bills. Obama Administration representatives have begun to testify before Congress on the President’s proposals, and the House and Senate Appropriations Committees are in the midst of drafting legislation establishing actual federal spending levels for FY 2015. As the FY 2015 budget process continues, lawmakers need to understand the impact that unpredictable funding and potential funding cuts will have on research programs, and they should be encouraged to acknowledge the value of biomedical research by maintaining the nation’s investment in NIH. Visit the ASH Advocacy Center (www.hematology.org/Advocacy) to read the latest news on NIH funding and to send a message in support of research funding to your elected officials. ASH Committee on Government Affairs Takes to Capitol Hill to Discuss Research Funding I n April, while Congress was beginning the FY 2015 budget process in earnest, the ASH Committee on Government Affairs conducted a Hill Day to discuss with congressional offices the importance of biomedical research and the need to protect NIH from funding cuts. These meetings with Congress are an important component of ASH’s advocacy efforts, providing an opportunity for Members of Congress and their staff to gain insight into issues of concern to hematologists. However, the Society needs the help of all members to bring issues important to the future of hematology to the attention of the U.S. Congress and other governmental agencies. ASH strongly encourages members to let the Government Relations, Practice, and Scientific Affairs staff know when you are in Washington, DC, and available to meet with your congressional delegation. ASH staff can assist by arranging appointments so that your voice is heard in the halls of Congress. You can also participate in the Society’s advocacy efforts by visiting the ASH Advocacy Center and by joining the ASH Grassroots Network. Contact ASH Legislative Advocacy Manager Tracy Roades at troades@hematology.org, or visit www. hematology.org/Advocacy for additional information on ASH’s advocacy efforts. ASH Provides Hematologists with Resources to Prepare for ICD-10 Conversion O n October 1, 2015, all health-care business transactions in the United States must convert from the use of the ninth version of the International Classification of Disease (ICD-9) to the 10th version (ICD-10). This change entails a complete restructuring of the diagnosis codes used by hematologists every day. The transition will take place on a single day, so there will not be any time to try the new diagnosis codes. ASH will be helping members to prepare for this transition, primarily by giving examples of the changes in hematology diagnosis coding. Twice a month, a new disease category will be released, comparing the ICD-10 codes with the ICD-9 codes. This review will provide clinicians with real examples so that they can understand the effect of the changes on their individual practices. Visit www.hematology. org/ICD10 to learn more about the ASH resources. If you have any questions about the implementation of ICD-10 or ASH’s resources, please contact Brian Whitman, ASH’s Senior Manager for Policy and Practice, at bwhitman@hematology.org or 202-776-0544. General Accountability Office Releases Report on Drug Shortages I n February, the General Accountability Office (GAO) issued a report titled, “Drug Shortages: Public Health Threat Continues, Despite Efforts to Help Ensure Product Availability,” which was called for in the Food and Drug Administration Safety and Innovation Act (FDASIA) of 2012. The report points to the combination of production lapses due to quality concerns and constrained manufacturing capacity as being the main cause of the large number of drug shortages experienced by the hematology community over the last several years. Encouragingly, the report states that the FDA has made progress over the past two years in preventing shortages by improving its recognition of and responsiveness to impending supply chain disruptions. GAO recommends that FDA further strengthen internal controls over data gathering that can alert the agency to a potential drug shortage and conduct periodic analyses to systematically assess drug shortage information so as to proactively identify drug shortage risk factors. Visit ASH’s drug shortage information page (www. hematology.org/DrugShortages) to access information about current hematologic drug shortages and to learn about ASH’s advocacy efforts and resources for physicians dealing with shortages. Update on the 2013 ASH Choosing Wisely Initiative Lisa K. Hicks, MD, MSc Staff Hematologist, St. Michael’s Hospital; Assistant Professor, University of Toronto; Chair of the Choosing Wisely Task Force Choosing Wisely is an initiative led by the ABIM Foundation in partnership with professional societies across the country (www.choosingwisely.org). This medical stewardship campaign aims to encourage physicians and patients to question the necessity of common medical tests and treatments. ASH released its first Choosing Wisely list at the ASH annual meeting in December 2013, and the full list and methodology that was used to develop the recommendations are available at www.hematology.org/choosingwisely. In brief, the ASH Choosing Wisely campaign suggests the following: 1) use the minimum number of red blood cell transfusion units to treat symptoms of anemia or to return a patient to a safe hemoglobin range (7 to 8 g/dl in stable, non-cardiac inpatients); 2) do not perform thrombophilia testing in patients with a provoked venous thromboembolic event (VTE); 3) do not use inferior vena cava (IVC) filters in the routine management of patients with a VTE; 4) do not use plasma to reverse the activity of vitamin K antagonists in non-emergency situations; and 5) limit CT surveillance for aggressive lymphoma in asymptomatic patients in complete remission. The Hematologist: ASH News and Reports After the release of the ASH Choosing Wisely List, the ASH Choosing Wisely Task Force was inundated with feedback from ASH members. The vast majority of feedback was positive and two major themes emerged. First, many members told us they want tools that will aid them in sharing the ASH Choosing Wisely message with their home institutions through teaching and continuing education sessions. In an effort to meet this request, ASH has developed a public-access Choosing Wisely slide deck. The slide deck summarizes the list, identifies the underlying evidence, and describes the methodology used to generate each of the ASH Choosing Wisely recommendations. The slide deck is available at www.hematology.org/choosingwisely. We encourage all members to use it, and pass it on. ASH is also developing a Choosing Wisely pocket guide that will be available on the same Web page. In addition to requests for tools to help disseminate the Choosing Wisely message, the Task Force received numerous emails suggesting that ASH produce a second Choosing Wisely list. In collaboration with the ABIM Foundation, ASH has already started working on a second list. Similar to the first ASH list, the second will be evidence-based and will prioritize tests and treatments that have limited benefit but a recognized risk of harm; the Task Force is aiming to release the second list at the annual meeting in December. The Choosing Wisely Campaign has a formal partnership with Consumer Reports and other consumer groups to both facilitate outreach to patients and direct dissemination to consumers. ASH has been working with Consumer Reports to produce an article geared toward patients, focusing on ASH’s recommendation to avoid routine use of IVC filters. We anticipate that this resource will be available in mid 2014. It will be posted on both the ASH Choosing Wisely landing page, (www.hematology.org/choosingwisely) and at www. consumerreports.org. The future challenges for the ASH Choosing Wisely Task Force are effective implementation and measurement of outcomes of the recommendations. How can we encourage the implementation of Choosing Wisely recommendations, and how can we determine if Choosing Wisely recommendations are leading to beneficial changes in patient care? These two related objectives are likely to be the most exacting aspects of this project. As yet, we on the ASH Choosing Wisely Task Force don’t have the answers to these questions, but we look forward to updating ASH membership as we tackle these issues. 7 Time to Retire the Old IMiD Mechanism of Action Slide Lu G, Middleton RE, Sun H, et al. The myeloma drug lenalidomide promotes cereblon-dependent destruction of Ikaros proteins. Science. 2014;343:305-309. Krönke J, Udeshi ND, Narla A, et al. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science. 2014;343:301-305. [Editor’s note: Data published in this paper were presented in abstract form by Dr. Krönke at the Late-Breaking Abstract Session during the 2013 ASH annual meeting in New Orleans.] T he publication of thalidomide’s remarkable and unanticipated anti-myeloma activity in Dr. Lu and colleagues showed the importance of modulating expression of IKZF1 and 19991 heralded the anti-cancer successes of its more potent analogs, lenalidomide IKZF3 in cell lines, demonstrating that cytotoxicity was observed only in cells in which and pomalidomide, over this past decade. The reemergence of these therapeutics, IKFZ1 and IKFZ3 expression was inhibited by lenalidomide. Additionally, both groups known as immunomodulatory drugs (IMiDs), as effective treatment for myeloma demonstrate that knockdown of either of these two targets is sufficient to induce and other hematologic malignancies is a bookend to thalidomide’s tragic beginnings, which cytotoxicity. Notably, Dr. Lu and colleagues observed resistance to lenalidomide-induced are inexorably linked to phocomelic infants born to women who took the drug to mitigate cytotoxicity in a number of myeloma cell lines. This unresponsiveness appears to be the emesis gravidarum. How thalidomide’s teratogenicity interdigitates with its biologic activity result of low-level expression of cereblon by the resistant cells, supporting the concept in various malignancies has been a focus of intense that the cytotoxic effects of lenalidomide are investigation for more than 20 years. Early illustrations cereblon-dependent through its action on Figure 1 Host Microenvironment depicting the mechanism of action of thalidomide expression of IKFZ1 and IKZF3 (Figure 2). IL-6 and lenalidomide highlighted autocrine and paracrine IGF-1 NF-kB VEGF interactions between myeloma cells and stromal cells A number of other purported mechanisms SDF-1a of the surrounding marrow microenvironment. An array for lenalidomide’s activity in multiple Inhibition of bFGF VCAM-1 MM cell adhesion of imaginative artistic techniques were used to propose myeloma have been reported, including ICAM-1 Inhibition to BMSC TNGa D of cytokine mechanisms of action that involve anti-angiogenic and down-regulation of expression of interferon VEGF C immunomodulatory pathways, induction of oxidative regulatory factor 4 (IRF4) and up-regulation circuits TGF-b stress, and modulation of cytokines, including downof expression of interleukin-2 (IL-2). The regulation of tumor necrosis factor-α and up-regulation authors of both papers tied the decrease in Thal/ImiDs of interleukin-2, which stimulates T-cell production IRF4 to the decrease of IKZF1 and IKZF3 Inhibition of (Figure 1). induced by lenalidomide, while Dr. Krönke et BFGF angiogenesis in al. demonstrated a marked decrease in IKZF1 VEGF BM milieu The first major breakthrough in understanding the and IKZF3 protein levels in T cells treated B mechanism of action of thalidomide and its derivatives with lenalidomide that was accompanied by a IL-2, IFNγ Enhanced host occurred when thalidomide was shown to bind to coincident increase in IL-2 expression. Based immune response cereblon, a critical component of an E3 ubiquitin ligase on these observations, it appears that these Apoptosis E complex (Figure 2).2 This report tied the teratogenic previously reported effects of lenalidomide (growth arrest) effects of thalidomide to inhibition of ubiquitination (IRF4 down-regulation and IL-2 up-regulation) T cells MM cell and toxic accumulation of proteins that led to cell are downstream effects of a lenalidomideA death. In the current reports published in Science, induced decrease in IKZF1 and IKZF3 Reprinted with permission © 2014 American Society of Clinical Oncology. All rights reserved. two independent groups have identified targets of (Figure 2). Richardson, P et al: Immunomodulatory analogs of thalidomide: an emerging new therapy in lenalidomide that account for its therapeutic effect myeloma. J Clin Oncol. Vol. 22 (16), 2004:3212-3214. in multiple myeloma. Dr. Gang Lu and colleagues Selective inhibition of transcription factors, from the laboratory of Dr. William Kaelin, Dana-Farber molecules otherwise considered undruggable, Cancer Institute, Boston, MA, and Dr. Jan Krönke and represents a novel therapeutic paradigm that Figure 2 colleagues from the laboratory of Dr. Benjamin Ebert, has emerged from the story of the relationship Brigham and Women’s Hospital, Boston, MA, report between IMiDs, cereblon, and IKZF1/IKZF3. that treatment with lenalidomide, and other agents in Data derived from these studies also provide this class, through interaction with cereblon, do not a foundation for exploiting these interactions inhibit, but rather selectively enhance the ubiquitination to design new therapeutics and to develop and degradation of Ikaros (IKZF1) and Aiolos (IKZF3), strategies for overcoming resistance to IMiDs. two members of the Ikaros family of zinc finger However, important issues remain unexplained. transcription factors important in B-cell development. The finding that the activity of IMiDs relies on This finding was confirmed through two orthogonal targeting transcription factors for proteasomal screening approaches. degradation seems incongruous with the highly effective clinical response observed when IMiDs In the work by Dr. Lu et al., 15,483 different genes were are combined with proteasomal inhibitors in the cloned into plasmids containing both firefly and renilla treatment of patients with myeloma. This paradox, luciferase and transfected into mammalian cells in order and the contrasting roles of Ikaros transcription to identify genes whose expression was affected by factors in different disease contexts (e.g., as tumor lenalidomide. By comparing the signal from these two suppressors in acute lymphocytic leukemia, and luciferases, the investigators were able to detect which as anti-neoplastic targets in myeloma), require of the 15,483 genes were transcribed into proteins better understanding. Further, while several and how levels of these proteins were affected by mechanisms of action have been proposed to lenalidomide. Expression of both IKZF1 and IKZF3 account for the therapeutic activity of lenalidomide decreased in the presence of lenalidomide. These in the treatment of del (5q) MDS, it is unclear findings were subsequently validated in two myeloma whether targeted ubiquitination and destruction of cell lines, MM1S and L363. specific transcription factors underlies the efficacy of lenalidomide in this disease. Using a different approach, Dr. Krönke et al. analyzed the proteome and ubiquitinome of MM1S cells after 1. Singhal S, Mehta J, Desikan R, et al. Antitumor treatment with lenalidomide. By using stable isotope activity of thalidomide in refractory multiple Immune modulators and myleoma. The small-molecule drugs thalidomide, labeling of amino acids in cell culture (SILAC) and myeloma. New Engl J Med. 1999;341:1565lenalidomide, and pomalidomide bind to the protein cereblon (CRBN), which mass spectroscopy, the authors identified a significant 1571. activates the enzymatic activity of the CRBN E3 ubiquitin ligase complex. lenalidomide-induced decrease in both IKZF1 and The transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) are modified with 2. Ito T, Ando H, Suzuki T, et al. Identification of IKZF3. The authors then went on to show that treatment ubiquitin (Ub) molecules, targeting them for proteolysis. This alters the function a primary target of thalidomide teratogenicity. with lenalidomide produced a decrease in protein (but of T cells and B cells, with a toxic outcome for multiple myeloma cells. Science. 2010; 327:1345-1350. not mRNA) levels of IKZF1 and IKZF3 in cells lines and From Stewart KA. How thalidomide works against cancer. Science. 2014;343:256-257. in primary patient samples. Reprinted with permission from AAAS. David Kurtz, MD, and Jason Gotlib, MD, MS Dr. Kurtz and Dr. Gotlib indicated no relevant conflicts of interest. 8 The Hematologist: ASH News and Reports Unlocking the Mysteries of AITL Odejide O, Weigert O, Lane AA, et al. A targeted mutational landscape of angioimmunoblastic T cell lymphoma. Blood. 2014;123:1293-1296. T hough angioimmunoblastic T-cell lymphoma (AITL) is a rare disease, it is one of the more common subtypes of peripheral T-cell lymphoma in the Western world. Patients typically present with advanced-stage disease and systemic symptoms, and associated autoimmune manifestations of the disease are common. Histologically, AITL is characterized by a proliferation of malignant T cells in a mixed background of polyclonal plasma cells, eosinophils, histiocytes, and B cells, often harboring the EpsteinBarr virus (EBV), in an expanded network of follicular dendritic cells with increased numbers of high endothelial venules. The pathogenesis of the disease is poorly understood and recurrent genetic abnormalities have not been systematically characterized. Using targeted exon capture and next-generation sequencing, Dr. Oreofe Odejide and colleagues in the laboratory of Dr. David Weinstock at Dana-Farber Cancer Institute in Boston examined a panel of 219 genes that were identified as mutated in other hematologic malignancies and correlated the findings with demographic and clinical data in 85 cases of AITL (Figure). AITL_ID TET2 DNMT3A IDH2 EP300 TP53 PCLO STAT3 ARID1B APC DST CREBBP JAK2 Age IPI ECOG_PS B_Symptoms LDH_Elevation 1. Yoo HY, Sung MK, Lee SH, et al. A recurrent inactivating mutation in RHOA GTPase in angioimmunoblastic T cell lymphoma. Nat Genet. 2014. Epub ahead of print. 2. Sakata-Yanagimoto M, Enami T, Yoshida K, et al. Somatic RHOA mutation in angioimmunoblastic T cell lymphoma. Nat Genet. 2014;46:171-175. 1 80 94 98 105 112 117 118 4 20 22 28 36 65 68 69 70 74 76 78 81 83 84 91 92 93 104 111 59 66 87 89 97 99 103 5 7 19 21 33 35 37 39 41 42 52 53 55 56 60 61 67 72 73 77 79 82 85 95 102 106 107 113 115 116 101 109 11 23 26 27 34 38 40 43 51 57 58 88 90 96 100 108 110 114 The median overall survival for patients was 18 months and was significantly inferior in patients over the age of 70. Mutations were identified in the coding regions of 80 genes, 34 of which were altered in two or more cases. The genetic changes did not differ according to the presence of EBV. Mutations in TET2 were present in 65 patients (76%), and 43 of 65 harbored two or three mutations in that gene. Twenty-eight patients (33%) had alterations in DNMT3A, all of which were seen in the presence of mutant TET2, and 17 patients (20%) had changes in IDH2, of which 15 also harbored TET2 mutations (Figure). Notably, alterations in TET2 and DNMT3A, which function as epigenetic modifiers of DNA, and in IDH2, a mitochondrial enzyme that when mutated produces the presumed oncometabolite 2-hydroxygluterate, are commonly found in myeloid neoplasms, and in contrast to many other lymphoid malignancies, alterations in TP53 were identified in only four cases. Given recent evidence suggesting that mutations in TET2 or DNMT3A may be derived from CD34+ progenitor cells, the investigators examined cellular subsets from a peripheral blood stem cell collection from a patient with AITL in remission whose tumor cells had been shown to harbor a TET2 mutation. That mutation was identified only in the lineage-negative CD34-/ CD38+ fraction of cells and not in the patient’s CD34+ positive cells or in cells with an immunophenotype similar to that of the patient’s AITL cells. The authors used the same experimental design to characterize the origin of the mutant cells in a patient with histology that overlapped AITL and peripheral T-cell lymphoma. In contrast to the prior case in which the mutation arose in a lineage-negative subpopulation, mutations in this case were acquired within lineage-committed progenitors. Additional investigation is needed to understand the origins of AITL. The prognosis of AITL is poor. The optimal management of the disease has not been well defined, and outcomes following anthracycline-based chemotherapy are dismal. Although a small minority of patients will achieve long-term disease control following stem cell transplantation, the vast majority of patients die of progressive disease. The work of Dr. Odejide and colleagues, identifying recurrent and overlapping mutations in TET2, DNMT3A, and IDH2, as well as recent reports demonstrating frequent alternations in RHOA, a small GTPase that regulates diverse biologic processes,1,2 raise the exciting possibility of novel therapeutic approaches in this disease. Studying drugs that work through epigenetic control over transcription, as well as investigating the efficacy of inhibitors of IDH2, may provide critically needed advances in the management of AITL. Key Age < 70 yrs Age >= 70 yrs IPI 1-2 IPI 3-5 ECOG PS 0-1 ECOG PS 2-3 no B Symptoms B Symptoms No LDH Elevation LDH Elevation Unknown Distribution of mutations in 85 cases of AITL. Common mutations and demographic and clinical factors across the cohort are shown. Each column represents a single patient. IPI = International Prognostic Index; ECOG PS = European Cooperative Oncology Group Performance Status. From Odejide O, Weigert O, Lane AA, et al. A targeted mutational landscape of angioimmunoblastic T cell lymphoma. Blood. 2014;123:1293-1296. Ann LaCasce, MD, MSc Dr. LaCasce indicated no relevant conflicts of interest. The Hematologist: ASH News and Reports 9 The Fate of Hematopoietic Nuclei: Major Roles for Lamins Before and After Marrow Release JAK-STAT Signaling Goes Bad in Primary Mediastinal Lymphoma Shin J, Spinler KR, Swift J, et al. Lamins regulate cell trafficking and lineage maturation of adult human hematopoietic cells. Proc Natl Acad Sci USA. 2013;110:18892-18897. Gunawardana J, Chan FC, Telenius A, et al. Nat Genet. 2014;46:329-335. M arrow hematopoietic cells comprise a semi-solid tissue that differentiates into various mature cell types that then populate the peripheral blood, a fluid tissue. The transition from marrow to blood requires that the mature blood cells acquire viscoelastic properties that allow them to pass through apertures as small as three microns as they traverse capillaries and narrow fenestrations in the marrow vascular endothelium, in the splenic red pulp, and in lymph nodes. Now, in an elegant series of experiments, Dr. Jae-Won Shin and colleagues in Dr. Dennis Discher’s laboratory at the University of Pennsylvania demonstrate that, depending upon hematopoietic lineage, expression of nuclear lamins play differing roles in determining both the fate of hematopoietic progenitor cells and the viscoelastic properties of mature nucleated blood cells. The two most common A-type lamins, lamin-A and lamin-C, are alternatively spliced products of one gene, while the two B-type lamins, B1 and B2, are products of separate genes. The lamin proteins form a mesh-like network, the nuclear lamina, which is located between and interacting with the inner membrane of the nuclear envelope and the peripheral heterochromatin of the nucleus.1 Nuclear lamins are also associated with nuclear scaffold proteins such as actin, with chromatin modifying proteins, and with transcription factors.1 These interactions can influence gene expression, nuclear shape, and nuclear deformability. Mutant A-type lamins are associated with myopathies, neuropathies, and progeria. In murine models, germ-line mutations in B-type lamins result in death early in embryogenesis, while mutant membrane receptors for B-type lamins are associated with progressive lymphopenia. 2 In humans, mutations in B-type lamin receptors cause the hyposegmentation of neutrophils that characterizes the Pelger–Huet anomaly.3 Dr. Shin and colleagues used mass spectrometry-calibrated intracellular flow cytometry to determine amounts and ratios of A-type and B-type lamins in normal human marrow and in nucleated blood cells. Using data from such analyses, the investigators developed a method for distinguishing early-stage hematopoietic cells from their mature progeny by plotting the content of A-type and B-type lamins versus the ratio of A-type:B-type lamins (Figure). In other experiments, they perturbed expression of lamins in hematopoietic progenitors using specific interfering RNAs or retinoic acid, which inhibits type-A lamin expression. They also tested nuclear deformability and chemotactic factorinduced cellular migration through three micron pores to simulate transit across capillary endothelium. Megakaryocytes contained the most total lamins, which increased with increasing ploidy, whereas the erythroid cells had the highest A-type:B-type lamin ratio, which increased with differentiation. The high lamin content of megakaryocytes and the nuclear rigidity associated with the high lamin A:lamin B ratio in erythroblasts make these marrow cell types least likely to migrate through the marrow vascular endothelium as they are the lineages that produce anucleate blood cells through proplatelet formation in the case of megakaryocytes and undergo nuclear extrusion prior to entering the circulation in the case of erythroblasts. Altered expression of A-type and B-type lamins also affected the differentiation of myeloid progenitor cells such that increased lamin-A expression and/or decreased lamin-B expression enhanced erythroid progenitor development but decreased granulocyte-monocyte progenitor growth. Lamin-A overexpression was also associated with increased megakaryocytic differentiation, while Figure decreased lamin-A expression inhibited erythroid progenitor differentiation and enhanced granulocyte-monocyte progenitor development. With decreased lamin-A:lamin-B ratios compared with erythroid nuclei and decreased lamin amounts compared with megakaryocytic nuclei, lymphocytic and myeloid nuclei had greater nuclear deformability, consistent with circulation of the mature nucleated cells in blood. During differentiation, the lamin-A:lamin-B ratio increased in granulocytic-monocytic cells, but their total lamin content decreased. Mature granulocytes-monocytes as well as lymphocytes readily migrated through three micron pores and softening their nuclei by decreasing the lamin-A to lamin-B ratio increased the migration rate. Log-log plot of intensity of nuclear lamins A + B versus ratio of lamin-A:lamin-B in human bone marrow and peripheral blood cells based on mean fluorescence intensity of A-type lamins and B-type lamins as quantified by mass spectrometrycalibrated intracellular flow cytometry. The dashed line demarcates cells residing in bone marrow (BM) in upper gray area and cells circulating in the peripheral blood (PB) in lower pink area. Abbreviations: CD34+,CD38-, cell population enriched in hematopoietic stem cells; CD34+,CD38+, cell population enriched in multipotent hematopoietic progenitors with limited proliferative capacity; MSC, mesenchymal stromal cells; MkP, megakaryocyte progenitors; MK, megakaryocytes; ProEry, proerythroblasts; lateEry, late-stage erythroblasts; BM-G and BM-M, bone marrow granulocytic and monocytic cells; PB GM, peripheral blood granulocytes and monocytes; T, T-lymphocytes; B, B-lymphocytes; RBC, erythrocytes; Plt, platelets. Shin J et al. Lamins regulate cell trafficking and lineage maturation of adult human hematopoietic cells. PNAS. 2013;110:18893. 10 Dr. Shin and colleagues demonstrated a role for nuclear lamins both in differentiation of marrow hematopoietic cells and in the nuclear flexibility required by mature nucleated blood cells to enter and remain in circulation. These results suggest that increased nuclear rigidity contributes both to the loss of circulating lymphocytes in lamin-B receptor-deficient mice2 and to the impaired chemotactic migration associated with the Pelger–Huet anomaly that is a consequence of mutant lamin B receptors in humans.4 The findings also suggest mechanisms that could account for the leukostasis observed in acute leukemias with high numbers of circulating blasts. The hypothesis in this case is that nuclei of the leukemic blast are more rigid than nuclei of normal mature granulocytic-monocyctic cells. Further, the high nuclear rigidity of circulating erythroblasts may contribute to the development of microvascular occlusions that complicate sickle cell anemia. 1. Zwerger M, Medalia O. Histochem Cell Biol. 2013;140:3-12. 2. Verhagen AM, de Graaf CA, Baldwin TM, et al. J Immunol. 2012;188:122-34. P rimary mediastinal B-cell lymphoma (PMBL) is distinct from other diffuse large B-cell types in its epidemiology, biology, and clinical course. Arising in the thymus, it is thought to originate in medullary B cells. It is characterized by a relatively increased incidence among young women, early local invasion, and a generally favorable prognosis after chemoimmunotherapy, often followed by consolidation radiotherapy. Failure of the initial therapy is uncommon, but it is often rapid when it does occur and extremely difficult to salvage. Gene-expression profiling has demonstrated strong similarities to nodular sclerosing Hodgkin lymphoma (HL), although no common underlying mutational events have previously been identified in more than a small number of cases. The pathogenesis of PMBL remains unclear, and for this reason, there is continuing investigation of its molecular characteristics. Led by Dr. Randy D. Gascoyne and Dr. Christian Steidl at the British Columbia Cancer Agency in Vancouver, Canada, this study has used whole-genome sequencing as the starting point for the identification of a previously unsuspected mutation target that is altered in a significant number of cases of both PMBL and HL. Initial whole-genome sequencing of material from two cases of PMBL, to a read depth of 60x and 52x, respectively, identified mutations in two negative regulators of the JAK-STAT signaling pathway, SOCS1 and PTPN1, in both cases. Subsequent sequencing of the whole transcriptome in another five cases and three PMBL cell lines revealed a high frequency of alterations in several regulators of the JAKSTAT pathway, with SOCS1 and PTPN1 being the most commonly affected. Next, the authors sequenced all 10 exons of PTPN1 in another 70 cases of PMBL and found a total of 18 variants in 17 out of 77 cases and an additional two variants in one of the three cell lines. Due to the possible overlapping biology with HL, they also sequenced PTPN1 in 30 cases and nine cell lines, finding another 12 variants among these. In clinical cases, the mutation rate of PTPN1 was 22 percent in PMBL and 20 percent in HL. Analysis of the PTP1B protein by immunohistochemistry suggested that the mutations generally resulted in reduced or completely absent expression, depending on the exact location and type of sequence variant. Investigating the functional consequences of these mutations showed that they impaired the phosphatase function of PTP1B, with higher levels of phospho-STAT6 seen in a reporter assay, suggesting constitutive upregulation of the signaling pathway through loss of inhibition. This hypothesis was supported by knock-down experiments using short-hairpin RNA to target PTPN1 transcripts in a wild-type HL cell line, which led to increased phosphorylation in several members of the JAK-STAT pathway and alterations in the gene-expression profile showing up-regulation of many STAT targets. Among these were oncogenes, including BCL-6, MYC, and JUN, and members of the gene family of ATP-binding cassette (ABC) transmembrane transporters, which are associated with resistance to chemotherapy. The evidence for de-regulation in the JAK-STAT pathway in PMBL is mounting steadily, and this study shows another way in which this pathologic process can come about. It has previously been difficult to identify a common pathogenetic pathway in PMBL, a disease with highly variable cytogenetic findings and few common alterations at the chromosomal level. However, the convergence of many different abnormalities seems to center on the JAK-STAT pathway and suggests a novel approach to treatment. The finding that some of the downstream targets of the mutations are mediators of drug resistance is also interesting, and larger studies are now needed to determine whether PTPN1 mutations convey an increased risk of primary treatment failure. 3. Gravemann S, Schnipper N, Meyer H, et al. Nucleus. 2010;1:179-89. 4. C unningham JM, Patnaik MM, Hammerschmidt DE, et al. Am J Hematol. 2009;84:116-119. mark koury, md peter johnson, MD Dr. Koury indicated no relevant conflicts of interest. Dr. Johnson indicated no relevant conflicts of interest. The Hematologist: ASH News and Reports Clots, Pumps, and Stockings – Do We Need to Update the Lower Extremity Dress Code? Genome Forensics Reveal Origins and the Mechanism that Drives t(12;21) Childhood B-ALL Kahn SR, Shapiro S, Wells PS, et al. Lancet. 2013. Epub ahead of print. Papaemmanuil E, Rapado I, Li Y, et al. RAG-mediated recombination is the predominant driver of oncogenic rearrangement in ETV6-RUNX1 acute lymphoblastic leukemia. Nat Genet. 2014;46:116-125. CLOTS (Clots in Legs Or sTockings after Stroke) Trials Collaboration, Dennis M, Sandercock P, et al. Lancet. 2013;382:516-524. E lastic compression stockings (ECSs) and intermittent pneumatic compression (IPC) devices have been used in a variety of medical settings, usually with the goal of restoring or maintaining normal venous return from the legs. Two recent publications provide important evidence about the effectiveness of these modalities in two different settings. The first study was a double-blinded, randomized, controlled trial of graduated ECSs (30 to 40 mm Hg at the ankle) versus placebo stockings (< 5 mm Hg compression at the ankle). The trial was designed to test the hypothesis that ECSs reduce the risk of post-thrombotic syndrome (PTS), a chronic complication of arm or leg deep-vein thrombosis (DVT). At 24 different North American centers, the “SOX” investigators enrolled 806 patients (mean age 55 years; 60% men) with a first, proximal leg DVT. Patients were included only if they 1) had a life expectancy > 6 months, 2) did not receive up-front thrombolytic therapy, and 3) had no contraindication to compression stockings. One year after randomization, the proportion of patients with PTS (as defined by validated, prespecified scoring systems) was 14 percent in the ECS arm and 13 percent in the “placebo stockings” arm. Similarly, overall quality of life (as measured by the SF-36 questionnaire) was not statistically different between the two groups. The second study was an unblinded, primary, VTE prevention trial, involving 2,876 patients (mean age 75 years, 52% women) from 105 hospitals in the United Kingdom who were both hospitalized for acute stroke (ischemic or hemorrhagic) and immobile. Patients in the CLOTS 3 trial were randomly assigned to receive either bilateral thigh-length IPC (n=1438) or no IPC (n=1438). Individuals with subarachnoid hemorrhage or leg problems (e.g., skin ulcers or severe edema) were excluded. Sequential compression was continued in the intervention group for 30 days or until the patient became independently mobile, was discharged, declined further use, or had adverse effects. All outcomes were evaluated 30 days after enrollment. The primary outcome (symptomatic or asymptomatic proximal DVT) occurred less commonly in the group assigned to IPC (9.6% vs. 14%; p=0.001). Symptomatic DVT, a key secondary endpoint, also occurred less frequently in the intervention group (4.6% vs. 6.3%; p=0.045). The rate of pulmonary embolism was not statistically different between the groups, but the numeric trend favored IPC (2.0% vs. 2.4%; p=0.453). IPC increased skin breaks (3.1% vs 1.4%, p=0.002). A strong, but statistically non-significant, trend toward lower 30-day mortality favored the IPC group (22% vs. 25%, p=0.057). Much can be learned from these two important prospective trials. The SOX trial is probably practice-changing. Until now, VTE experts have recommended that most patients with proximal leg DVT wear ECSs (30 - 40 mm Hg at the ankles) because two previous studies1,2 had indicated that ECS use could decrease the risk of PTS by 50 percent. However, neither of these often cited studies was blinded. The PTS score, which is the key outcome measure of all these trials, depends heavily on subjective assessments by both clinicians and patients; thus the possibility of bias in unblinded comparisons is significant. By using “placebo” stockings in the comparator arm, the SOX investigators eliminated this kind of bias and found no benefit from ECSs. More evidence is needed about how we might reduce the risk of PTS before it occurs. While ECSs may still be worth trying as a palliative measure in the 10 to 15 percent of patients who develop moderate to severe PTS, the SOX trial suggests we will need to look beyond ECSs if we hope to prevent PTS in the first place. The CLOTS-3 trial is by far the largest and most rigorously designed study that shows that IPC devices can substantially reduce the risk of VTE in a non-surgical population of hospitalized patients. Hematologists are often asked to comment on the risks and benefits of VTE prevention. For some populations (e.g., patients admitted for acute leukemia or hematopoietic cell transplantation), pharmacologic modalities not only are costly but also probably carry a non-trivial risk of bleeding. The CLOTS-3 study, along with a recent meta-analysis,3 provides high-quality evidence that IPC, when properly applied, can provide substantial protection against VTE. A t times, some of the most fundamental issues in human biology have appeared inaccessible to direct experimental investigation and have consequently been relegated to study using model systems. The origin of childhood precursor-B cell acute lymphocytic leukemia (B-ALL) was included in that group until the discovery 15 years ago of the prenatal origin of the t(12;21) translocation that generates the ETV6-RUNX1 (formerly TEL-AML1) fusion gene found in 25 percent of childhood ALL. In a succession of studies using blood samples from twin pairs, neonatal blood spots from archived Guthrie cards, and stored cord blood units, investigators showed that the ETV6-RUNX1 fusion gene developed in utero at an astounding frequency of one percent.1 Clearly, 1 percent of children do not develop B-ALL. Thus, events acting in concert with t(12;21) are necessary for leukemogenesis. During the Ham-Wasserman Lecture at the 2009 ASH annual meeting, Professor Mel Greaves, who had pioneered work in this area, made a compelling case for incorporating Darwinian evolutionary principles into a conceptual framework for understanding ALL leukemogenesis.1 But at the time, the genetic events underlying clonal evolution of ETV6-RUNX1 expressing cells were speculative. Was the process merely stochastic, or was it connected mechanistically to t(12;21)? The expanding use of genome-scale technology over the past several years only added to the confusion by identifying many seemingly random genetic abnormalities. Now, the current study, led by Prof. Greaves from the Institute for Cancer Research in London and by Professor Peter Campbell from the University of Cambridge, Cambridge, UK, reports identification of the mechanism that underlies clonal expansion in B-ALL expressing ETV6-RUNX1. In their detailed genetic analyses, Dr. Papaemmanuil and colleagues studied samples from 57 ETV6-RUNX1 positive ALL patients. Using both exome and low-coverage whole-genome sequencing, they found an average of 11 somatically acquired structural variations per case, mostly deletions, with half being highly recurrent. Notably, deletions mostly affected genes involved in B-cell differentiation. When the authors analyzed the nucleotide sequence of the structural variants, they observed a striking frequency of recombination signal sequence (RSS) motifs spanning breakpoint junctions. This finding suggested that recombination-activating endonucleases (RAG)-1 and -2 were involved in creation of the structural variants. RAG-1 and -2 are essential components of the process involved in generating antibody and T-cell receptor diversity through mediation of VDJ gene recombination. This process involves DNA cleavage and the addition of nontemplate nucleotide bases, which incidentally leaves a genomic fingerprint of RAG activity. The authors found that 40 percent of the analyzed structural variants had RAG recognition sequence (RSS) motifs and that 70 percent had inserted non-template sequence. Two other intriguing findings were the unexpectedly high incidence of structural variants that localized within promoter and enhancer regions, many of them coinciding with RSS motifs, and identification of mutations in two previously unrecognized tumor suppressor genes, ATF7IP and MGA. Interpreted as a whole, the mutational pattern suggests successive inactivation of genes that would otherwise promote differentiation, stalling the ETV-RUNX1 expressing cells in a stage of early B-lineage differentiation. Sustained high RAG activity would then drive clonal evolution, converting a pre-leukemic cell into a malignant clone. This rigorous study provides a detailed genetic fingerprint of ETV6-RUNX1 B-ALL, revealing RAG-mediated deletions as the driver of leukemogenesis. The contribution of this process to leukemogenesis in other subtypes of ALL will be the subject of future studies. The hope in understanding leukemic evolution is, of course, the possibility of translation of that understanding into development of effective treatment. Knowing the mechanisms at work, and the genomic fingerprints that signal progression to full-blown disease versus regression, might not only allow for development of targeted therapy, but perhaps suggest an approach to preemptive intervention. This study is also noteworthy for identification of novel immunosuppressor genes and for the interwoven use of existing databases with innovative bioinformatics. The authors’ genome forensics focused on t(12;21) ALL, but their experimental design strategies may help gain insight into other pediatric and adult malignancies. 1. Greaves M. Darwin and evolutionary tales in leukemia. Hematology Am Soc Hematol Educ Program. 2009;2009:3-12. 1. Prandoni P, Lensing AWA, Prins MH, et al. Below-knee elastic compression stockings to prevent the post-thrombotic syndrome: a randomized, controlled trial. Ann Intern Med. 2004;141:249-256. 2. Brandjes DPM, Büller HR, Heijboer H, et al. Randomised trial of effect of compression stockings in patients with symptomatic proximal-vein thrombosis. Lancet. 1997;349:759-762. 3. Ho KM, Tan JA. Stratified meta-analysis of intermittent pneumatic compression of the lower limbs to prevent venous thromboembolism in hospitalized patients. Circulation. 2013;128:1003-1020. david garcia, MD Kelsie J. Storm, MD, and Peter Kurre, MD Dr. Garcia indicated no relevant conflicts of interest. Dr. Storm and Dr. Kurre indicated no relevant conflicts of interest. The Hematologist: ASH News and Reports 11 Lessons From the Liver: Rebalanced Hemostasis in Immune Thrombocytopenia Another Reason to Eat More Apples and Whole Grain: How Inflammation and Hematopoiesis are Affected by Dietary Fiber and GI Microbes Kim WH, Park JB, Jung CW, et al. Rebalanced hemostasis in patients with idiopathic thrombocytopenic purpura. Platelets. 2014. Epub ahead of print. Trumpette A, Gollwitzer ES, Yadava K, et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med. 2014;20:159-166. C O ommon laboratory tests of the hemostatic system, such as the platelet count, activated partial thromboplastin time (aPTT), and prothrombin time (PT), are often abnormal in patients with liver disease. If interpreted in isolation, these laboratory abnormalities appear to indicate a tendency for the patient to bleed. Consequently, clinicians often transfuse blood products in an attempt to normalize these laboratory abnormalities, especially before invasive procedures. In recent years, there has been an increasing understanding of a “rebalancing” of the hemostatic system in liver disease, whereby hemostatic changes that promote bleeding can be counterbalanced by hemostatic changes that promote thrombosis.1 Thus, the platelet count, aPTT, and PT, although commonly ordered, fail to reflect the globally rebalanced hemostasis of liver disease, which can lead to unnecessary and possibly harmful treatments. Indeed, preoperative “correction” of these laboratory abnormalities does not necessarily reduce bleeding and may actually promote it. Immune thrombocytopenia (ITP) is another disorder in which a laboratory abnormality (i.e., the platelet count) does not accurately predict the risk of bleeding, especially for the individual patient. A singular focus on the platelet count in ITP can cause a clinician to make the treatment for ITP worse than the disease itself, especially for childhood ITP. Such unnecessary therapy and resulting iatrogenic morbidity has been called “medical nemesis.”2 To decrease medical nemesis in ITP, it is important to begin to understand why some patients do not bleed despite severe thrombocytopenia and why severe hemorrhage can occur despite treatment to raise the platelet count. One possible explanation could be that there are hemostatic differences beyond the platelet count that predispose to severe bleeding. Dr. Won Ho Kim and colleagues from Sungkyunkwan University School of Medicine, Seoul, Republic of Korea, sought evidence for a rebalancing of hemostasis in ITP, similar to that proposed for liver disease, which could explain the frequent disconnect between the platelet count and the occurrence of bleeding. They built upon prior observations that a global assessment of hemostasis (thromboelastography) might predict bleeding in ITP3 and that the reported decrease in von Willebrand factor (vWF) cleaving protease activity in ITP4 could conceivably mitigate the effects of thrombocytopenia. Dr. Kim and colleagues measured vWF antigen levels and performed rotational thromboelastography in 20 adults with ITP and followed them for six months for the occurrence of bleeding. The mean vWF antigen level was 163 percent (S.D. 80%), and eight patients (40%) had elevated vWF antigen levels (218%, S.D. 104%) when adjusted for their blood group type. Thromboelastography parameters were within the normal range for most patients. A subgroup analysis compared patients with normal versus elevated vWF. Those with elevated vWF were older and had longer duration of ITP. The clot formation time (CFT) was shorter, and the α-angle was larger in those with elevated vWF. The CFT and α-angle are sensitive to platelet number and function and fibrinogen level and polymerization. So, higher vWF levels were associated with better thromboelastography values compared with lower vWF values despite a similar platelet count. Severe bleeding did not occur in any patient, but four patients had minor bleeding (3 in the normal vWF group). The study by Dr. Kim and colleagues was small and many aspects of the hemostatic system were not measured, but they did document evidence for “rebalanced hemostasis” in ITP. The main finding was an association between high vWF antigen levels (in 8/20 patients) and improved thromboelastographic measurements that might account for a decreased tendency toward bleeding. Although these results are intriguing, they cannot yet tenably inform clinical practice. Hopefully, further research can more completely explain why some patients with ITP bleed and some do not. Possible explanations include the effects of the autoantibody on platelet function and a rebalance of hemostasis analogous to that of liver disease. Whatever the answers, we clearly need better laboratory assessments to understand the global hemostatic consequences of ITP, which would also help to reduce medical nemesis in this challenging disease. To examine the role that dietary fiber plays in modulating AAI, mice were fed a standard diet containing 4 percent fiber or a low-fiber diet containing 0.3 percent fiber and exposed to house dust mites. Compared with mice fed the low fiber diet, mice fed the higher-fiber diet had less lung eosinophil and lymphocyte infiltration, reduced production of cytokines IL-4, IL-5, IL-13, IL17A, lower levels of IgE, and IgG1 antibodies against the dust mite. Mice fed a high-fiber diet had less severe AAI in response to methacholine, and lung DCs were less activated as shown by lower expression of CD40, CD80, PDL-1, and PDL-2. Complementary studies confirmed these findings in mice that were fed a diet supplemented with the fermentable fiber pectin (a substance enriched in apples). Dietary fiber changed the gut and lung microbiota by enhancing bacterial diversity, especially in the proportion of bacteroidaceae and bifidobacteriaceae, both potent fermenters of fiber into SFCAs. Both serum and cecal acetate and propionate levels were higher in the high-fiber fed animals. To show that increased SCFAs mediate the protective effect of fiber against inflammation, mice were fed acetate or propionate. One day after mice were given an intranasal house mite challenge, inflammation was similar in control- and propionatefed animals, but after two to four days, eosinophils and cytokine levels in the lung were reduced in SCFAs-treated mice. This protective effect was dependent on G-protein coupled receptor 41 (also called free fatty acid receptor 3). There was impaired TH2 differentiation and decreased dendritic cell (DC) activation in SCFAs-treated mice four days after the challenge. Other experiments showed that propionate enhanced hematopoiesis, increasing both common DCs and macrophage DCs in the bone marrow. This study demonstrates the impact that dietary fermentable fibers have on the intestinal microbiome, allergic/immune reactions, and bone marrow function. More than 40 years ago, studies in germ-free or completely decontaminated mice showed that the absence or elimination of the intestinal microflora prevented the development of delayed-type acute graft-versus-host disease (GVHD) in mismatched bone marrow transplant models, putatively because of the experimentally induced absence of gastrointestinal anaerobic bacteria in these animals.2-4 Recently, interest in gut microbes and GVHD has re-emerged.5-7 Antibiotics, diet, immunosuppressive agents, radiation, and chemotherapy treatment alter the intestinal microbial milieu and influence the barrier functions of the bowel, and the effects on GVHD of interventions ranging from gut bacterial decontamination, to protective environments, to the addition of certain bacteria into the gut are the subjects of ongoing investigation. Prof. Burkitt’s hypothesis has proven prescient, and continued study of the relationship between dietary fiber intake, the intestinal microbiome, systemic immuno/inflammatory processes, and bone marrow function is warranted to further explore the wide spectrum of associated disease pathophysiology and to identify new therapeutic targets. 1. Burkitt DP, Walker AR, and Painter NS. Effect of dietary fibre on stools and the transittimes, and its role in the causation of disease. Lancet. 1972;2:1409-1412. 2. Jones JM, Wilson R, Bealmear PM. Mortality and gross pathology of secondary disease in germfree mouse radiation chimeras. Radiat Res. 1971;45;577-588. 1. Lisman T, Porte RJ. Rebalanced hemostasis in patients with liver disease: evidence and clinical consequences. Blood. 2010;116:878-885. 3. van Bekkum DW, Roodenburg J, Heidt PJ, et al. Mitigation of secondary disease of allogeneic mouse radiation chimeras by modification of the intestinal microflora. J Natl Cancer Inst. 1974;52:401-404. 2. Lilleyman J. Medical nemesis and childhood ITP. Br J Haematol. 2003;123:586-589. 4. Beelen DW, Haralambie E, Brandt H, et al. Evidence that sustained growth suppression of intestinal anaerobic bacteria reduces the risk of acute graft-versus-host disease after sibling marrow transplantation. Blood.1992;80:2668-2676. 3. Gunduz E, Akay OM, Bal C, et al. Can thrombelastography be a new tool to assess bleeding risk in patients with idiopathic thrombocytopenic purpura? Platelets. 2011;22:516-520. 4. Moore JC, Hayward CPM, Warkentin TE, et al. Decreased von Willebrand factor protease activity associated with thrombocytopenic disorders. Blood. 2001;98:1842-1846. 12 ne day, sometime in the 1970s, when I was a medical student attending Medicine Grand Rounds, I remember hearing Professor Denis Burkitt extol the wonders of dietary fiber, arguing persuasively that, compared with Westerners, Africans experience a lower incidence of diseases ranging from colon cancer to hemorrhoids because their diet is relatively fiber-rich. The mechanisms that accounted for the beneficial effects of high dietary fiber were unclear at the time, and the simple concept of more rapid stool transit time was invoked to explain the difference. 1 Subsequent investigation, however, has uncovered an intricate relationship between dietary fiber, the microbes in our gut, and both intestinal and systemic immuno/inflammatory processes. Dietary fibers are complex carbohydrates that can be fermented by certain species of gut bacteria, leading to production of bioactive products including short-chain fatty acids (SCFAs). SCFAs serve as an energy source for certain bacteria, but they also provide fuel for intestinal epithelial cells. Available evidence suggests that their effects are exerted through a surprisingly diverse range of mechanisms including engagement of G-protein coupled receptors and inhibition of histone deacetylase activity. And SCFAs such as acetate, propionate, and butyrate are not confined to the gut, being disseminated diffusely after entering the circulatory system and thereby becoming available to participate in systemic processes. Now, from Dr. Aurélien Trompette and colleagues at the University of Lausanne, in Lausanne, Switzerland, we learn how generation of SCFAs by gut microbiota metabolism both ameliorates allergic airway inflammation (AAI) and drives hematopoiesis. 5. Rezvani AR and Storb RF. Prevention of graft-vs.-host disease. Expert Opin Pharmacother. 2012;13:1737–1750. 6. Jenq RR, Ubeda C, Taur Y, et al. Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation. J Exp Med. 2012;209:903-911. 7. Bryson JS, Brandon JA, Jennings CD, et al. A gut feeling about murine syngeneic GVHD. Chimerism. 2011;2:58-60. charles T. Quinn, MD, ms Gregory M. Vercellotti, MD Dr. Quinn indicated no relevant conflicts of interest. Dr. Vercellotti indicated no relevant conflicts of interest. The Hematologist: ASH News and Reports Clinical Trials Corner Not Just for GIST: A New FLT3 Inhibitor Active in Kinase Domain Mutated AML Study Title: A Phase II Study of Crenolanib in Relapsed/Refractory Acute Myeloid Leukemia Patients With FLT3 Activating Mutations Clinical Trials.gov Identifier: NCT01657682 Coordinator: Arog Pharmaceuticals LLC Participating Center: MD Anderson Cancer Center Accrual Goal: 41 Rationale: AML patients with FLT3 internal tandem duplications (ITD) have a poor prognosis. Several FLT3 TKIs have been studied in clinical trials, and the results have been disappointing to date because of lack of efficacy and treatment-related toxicity (Serve H et al. J Clin Oncol. 2013;31:3110-3118). Crenolanib was initially found to have activity against the imatinib-resistant PDGFRA-mutant kinases that drive gastrointestinal stromal tumors (GIST) (Heinrich MC at al. Clin Cancer Res. 2012;18:4375-4384). In addition, it is not only a potent inhibitor of FLT-ITD, but it is also active against FLT3 with gain-of-function mutations in the tyrosine kinase domain (TKD), including D835H/Y. Comment: About 30 percent of AML patients have FLT3- ITD mutations, and another 8 percent have FLT3-TKD mutations. Studies of sorafenib, one of the early TKIs tried in AML patients with FLT3-ITD, did not show appreciable benefit despite early reduction in blasts (Serve H et al. J Clin Oncol. 2013;31:3110-3118). This ultimate lack of efficacy was explained by near uniform development of drug resistance, by 72 days on average, attributed to emergence of a clone with the D835 mutation in the kinase domain of FLT3 (Man CH et al. Blood. 2012;119:5133-5143). Subsequent trials were conducted with midostaurin (Stone RM et al. Leukemia. 2012;26:2061-2068) and quizartinib (Cortes JE et al. J Clin Oncol. 2013;31:3681-3687). The latter is believed to be one of the most active clinical agents. However, these drugs are not active against ASH News and Reports – Pamela S. Becker, MD, PhD Dr. Becker indicated no relevant conflicts of interest. Study Design: This is a phase II study of a new Fmslike tyrosine kinase 3 (FLT3) inhibitor (crenolanib) in relapsed or refractory acute myeloid leukemia (AML). The study includes two cohorts: one with relapsed/refractory AML without prior FLT3 tyrosine kinase inhibitor (TKI) treatment, and one with relapsed/refractory AML with prior FLT3 TKI treatment. The primary endpoints are response rate after the first 28-day cycle and at best response and safety. There is a stopping rule that will be applied if there is more than a 95 percent chance that the toxicity rate exceeds 30 percent, with toxicity defined as a grade 4 or greater non-hematologic adverse event. The secondary endpoints are duration of clinical response and progression-free and overall survival. Pharmacodynamic markers, including phospho-FLT3, will be assayed in blood and marrow blast samples, and pharmacokinetic studies will be performed to determine the relationship of drug concentration to response and toxicity. Patients in each cohort receive the same dose of crenolanib besylate (200 mg/m2/day) divided into three daily doses, preferably every eight hours, taken orally at least 30 minutes before or after a meal. Concurrent hydroxyurea (maximum 5g total daily dose for 14 days) is permitted during the first 28 days of study therapy. The Hematologist: cells with FLT3-TKD activating mutations. A recent publication showed efficacy of crenolanib in laboratory studies of drug-resistant FLT-TKD-mutated leukemia cells (Zimmerman EI et al. Blood. 2013;122:3607-3615). The current clinical trial will assess the efficacy of crenolanib in two groups of patients: those without prior treatment with TKIs and those who developed resistance after TKI therapy. The two-cohort design should provide data about whether the activity against FLT3-TKD mutants observed in pre-clinical studies translates to the clinic and whether use of the inhibitor in naïve patients prevents development of drug resistance. Rivaroxaban for the Treatment of HIT: Hit or Miss? Study Title: Rivaroxaban for Treatment of Patients with Suspected or Confirmed Heparin-Induced Thrombocytopenia ClinicalTrials.gov Identifier: NCT01598168 Sponsor and Collaborator: McMaster University Participating Centers: 7 medical centers in Canada Accrual Goal: 200 Study Design: This is a prospective, multicenter, singlearm cohort study (Linkins LA et al. J Thromb Thrombolysis. 2014. Epub ahead of print). The target population is adult patients with an intermediate or high clinical probability (4T score ≥ 4) of heparin-induced thrombocytopenia (HIT). Patients with mechanical heart valves, severe renal insufficiency (CrCl <30 mL/min), or moderate or severe hepatic disease (Child-Pugh B and C) are excluded. All enrolled subjects are initially given 15 mg of rivaroxaban twice daily while awaiting the results of HIT laboratory testing. If laboratory testing is negative, rivaroxaban is discontinued. In patients who test positive for HIT, rivaroxaban is continued for 30 days. The dose is changed to 20 mg daily after 21 days in patients with thrombosis, and, in patients without thrombosis, the dose is changed at the time of platelet count recovery (defined as platelet count ≥ 150 x 109/L or recovery to platelet count at the time heparin was initiated). The primary outcome is new symptomatic venous or arterial thromboembolism at day 30. Secondary endpoints include major bleeding and time to platelet count recovery. In addition to local testing, serum from all subjects will be analyzed for platelet activating antibodies in a central laboratory (McMaster Platelet Immunology Laboratory) using a serotonin release assay (SRA). Rationale: Drugs licensed for the treatment of HIT (e.g., argatroban, danaparoid, lepirudin) are associated with important drawbacks, including parenteral administration, need for intensive laboratory monitoring, limited availability (in the case of danaparoid and lepirudin), high cost, and incomplete efficacy. These agents decrease thromboembolism by 50 to 65 percent but do not reduce HIT-associated amputation or mortality. Transitioning from argatroban to warfarin is challenging because of the effect of argatroban on the INR and added length of hospital stay. Rivaroxaban, a direct inhibitor of free and clot-bound factor Xa, is a promising agent for the management of HIT that may overcome some of these limitations. It is administered orally as a fixed dose, is less costly than parenteral agents, does not require routine laboratory monitoring, does not cross-react with HIT antibodies in vitro (Walenga JM et al. Br J Haematol. 2008;143:92-99), and has an established efficacy and safety profile in the management of other thromboembolic disorders. Comment: There is an urgent need for novel therapeutics in HIT that improve upon the efficacy, safety, convenience, and cost-effectiveness of existing options. Whether one or more of the target-specific oral anticoagulants will be able to fill this niche is unproven. This study will provide valuable preliminary information but, due to two important limitations in its design, will not definitively address the role of rivaroxaban in the management of HIT. First, the study lacks a control group against which rivaroxaban may be compared. Ideally, patients would have been allocated to receive either rivaroxaban or an active comparator (e.g., argatroban). However, randomized controlled trials have proven difficult to conduct in HIT; only two such trials have been published. A trial of danaparoid versus dextran 70 was completed in the 1990s prior to the advent of approved agents (Chong BH et al. Thromb Haemost. 2001;86:1170-1175). A more recent trial of desirudin versus argatroban was terminated early due to poor accrual after only 16 subjects had enrolled (Boyce SW et al. Am J Ther. 2011;18:1422). Comparisons between the rivaroxaban cohort and the pivotal single-arm studies that led to approval of argatroban and lepirudin are unlikely to be meaningful because of differences in patient population, outcome assessment, and adjunctive care. Second, it is likely that the majority of patients enrolled in the study will not have serologically confirmed HIT. In a recent meta-analysis, HIT was corroborated using a specific washed platelet assay (e.g., SRA) in only 23 percent of patients with suspected HIT and a 4T score ≥ 4 (Cuker A et al. Blood. 2012;120:4160-4167). Thus, it can be anticipated that 50 or fewer of the 200 subjects enrolled in the study will have “true” serologic HIT. Whether this number will be sufficient for meaningful assessment of rivaroxaban for the treatment of HIT remains to be determined. The aforementioned limitations notwithstanding, this study represents an important step forward in the investigation of HIT. It places renewed focus on a potentially devastating orphan disease with limited treatment options and no new FDA-approved therapies since argatroban received licensure 14 years ago. Despite lack of a control arm, the quality of evidence obtained from this study will undoubtedly exceed that derived from retrospective analyses cited to support off-label use of fondaparinux and bivalirudin for HIT. If results are promising, this study should pave the way for an international, randomized, controlled trial to definitely address the role of rivaroxaban for treatment of HIT. – Adam Cuker, MD, MS Dr. Cuker indicated no relevant conflicts of interest. 13 P r o f i l e s in H e m a t o l o g y Hematology as a Journey John W. Adamson, MD Clinical Professor of Medicine, Hematology/Oncology, University of California, San Diego Awed by the physician who removed my tonsils (probably unnecessarily), I have wanted to be a doctor since the age of three. Not that there weren’t doubts along the way, as years later as a student at the University of California, Berkeley, I envisioned the road (medical school, internship, residency) that I would have to travel to reach that goal. For a time, I considered following my father’s lead, but he, a high school English teacher, straightened me out. “Don’t be stupid!” he said. “Being a teacher is fine, but you can teach at whatever you do.” Of course, he was right, and in the end, I had it both ways, as I became a physician/teacher. When it came time to apply to medical school in 1958, there were the usual options for someone growing up in California: Stanford, the University of California, San Francisco, the University of Southern California, and the newly minted University of California, Los Angeles (UCLA). For whatever reason (likely intuitive), I chose UCLA. This decision led to my first airplane ride when I moved from Berkeley to Los Angeles. The experience at UCLA was eye-opening. Most of the lectures were given by young faculty who were deeply invested in their own research, so, predictably, part of the class complained about the emphasis on science and the others enjoyed the newness of it all. As for internships, I didn’t apply very widely, and there wasn’t anything like today’s computerized system that “matches” applicants with a particular training program. Fortuitously, I wound up in Seattle at the University of Washington where I found myself among a group of remarkable biomedical researchers including Earl Davie, Clem Finch, and the nephrologist Belding Scribner, who was pioneering hemodialysis as an approach to treating kidney failure. Toward the end of my second year of residency in 1964, I was asked if I would be interested in becoming a hematology fellow. I don’t know why, but someone had given my name to Clem, who was head of the Division of Hematology at the time, and he approached me personally. After some thought, I accepted the invitation and “shorttracked,” entering fellowship after two years as a resident. I reasoned that if I didn’t like hematology, I would complete my third year of internal medicine training and go into practice. Fellowship programs were so much less formulaic then. I spent most of the first two years (1964-1966) in the lab and fell in love with it. I was assigned to help develop a new assay for the elusive erythropoietic stimulating factor (i.e., erythropoietin or Epo), and I plunged in. We started with samples from normal subjects and then moved to patient samples. The bioassay wasn’t sensitive enough to reliably measure Epo activity in normal serum, so we had to concentrate urine to detect a signal. These experiments led to the discovery that Epo levels were consistently low in patients with polycythemia vera, and we went on to use the assay to analyze Epo levels in a number of polycythemic states. Things were going smoothly until 1967, when it became clear that I was going to be drafted into the military. Coming to my support, Clem called friends at the National Institutes of Health (NIH) who informed him that there was an unexpected opening in hematology in the Clinical Pathology Department. I literally flew out that night, interviewed the next day, and, thankfully, was offered the opportunity to apply for the position. When I returned to Seattle, I found waiting for me a draft notice stating that I was to report to the Presidio of San Francisco on July 5. Now it was a race to see what would come first: the offer of a commission in the Public Health Service or July 5 and the Army. I was going somewhere, but I just didn’t know where. Mercifully, the offer to join the commissioned corps arrived about a week before July 5, and I was off to Bethesda, Maryland. NIH was the center of biomedical research at that time, and a second postdoctoral experience there was a watershed event in my career. It was at NIH that I learned the basics of cell culture and the physical separation of cells, tools that made 14 feasible the study of subpopulations of hematopoietic cells. And through the team of James Till and Ernie McCulloch at the Ontario Cancer Institute (OCI) in Toronto, I was exposed to novel concepts in the new world of experimental hematology, as Till and McCulloch had made seminal observations about the nature and function of stem cells and about clonality and clonal evolution. Ernie was very supportive, and I had the opportunity to visit the OCI on several occasions at a time when colony-forming assays were being developed. When my time at NIH was up, Bob Petersdorf, chair of Medicine at Washington, offered me the opportunity to return, and I joined the faculty in 1969. I now had new tools to apply to the lines of investigation that had been started earlier. My work in the lab in Seattle linked two areas: the clonal nature of the myeloproliferative neoplasms (in collaboration with Phil Fialkow) and the potential value of Epo as a therapeutic agent (in collaboration with my close friend, Joe Eschbach). These two areas, along with ongoing studies of iron physiology led by Clem, dominated the next 20 years of my academic life in Seattle. Bev Mitchell joined the lab early on and became a good friend. It was an exciting time. Joe and I had developed a large animal model of renal failure and convincingly demonstrated that plasma that was rich in Epo could reverse the accompanying anemia. As a result, scientists from Amgen, then a small startup company, contacted us to see if their recombinant human Epo would work in our animal model. We never got there! Rather, we quickly initiated the first clinical trials of Epo, in which we assessed response in transfusion-dependent patients on dialysis; it was amazing to see something work so well! To extend our work on clonal disorders, we developed a model of G-6-PD heterozygosity in the cat – a model that Jan Abkowitz, then a fellow and currently ASH’s immediate past president – exploited brilliantly. Ken Kaushansky, also a fellow at the time, introduced molecular biology to the division in studies of hematopoietic growth factors, their receptors, and their biology. Ken’s work in this area culminated in his cloning and expression of the gene that encodes thrombopoietin (Tpo), Epo’s little sister. My years in Seattle were interrupted by another important event — a year on sabbatical with (now) Sir David Weatherall at Oxford. The Weatherall lab was studying the regulation of globin chain synthesis in an attempt to better understand the pathophysiology of hemoglobinopathies. While I probably disappointed David in how little I accomplished, living abroad and watching really good minds in action in a resource-restricted environment was revelatory. Through that experience, I made many friends and adopted a view of the world that I don’t believe I would have had it not been for the experience of living outside of the United States. I also came to like football, as it’s defined on the rest of the planet. In 1989, I accepted a new challenge, moving to New York City and the New York Blood Center with a goal of expanding the research programs there. Fortunately, a very talented husband-and-wife team of scientists, Anna Rita and Giovanni Migliaccio, accompanied me, and they sustained and grew the laboratory by encouraging and supporting the work of visiting fellows and scientists. New York was a challenge, but very exciting. In 1998, I was recruited to the Blood Research Institute of the BloodCenter of Wisconsin in Milwaukee, which Dick Aster had led for many years. The Institute was home to a group of extremely bright scientists including Dick, Peter Newman, Bob Montgomery, and Hardy Weiler. My time at the BloodCenter provided one of the best experiences of my life, and with the support of the scientists and the Institute’s Board, we were able to recruit a number of outstanding young investigators who have gone on to success in their own right. The final move, in 2007, completed the circle, bringing me back to California and to the University of California, San Diego (UCSD), where I rejoined Ken Kaushansky. At that time, Ken was chair of the Department of Medicine and (with some irony) my ultimate boss. The move to UCSD was in support of my wife’s career, and she now heads the Center for Hemophilia and Thrombosis here. With Sandy Shattil’s support, I try to do what I can to remind people (especially the fellows) that there is such a thing as benign hematology and that the study of blood should be a passion, not a pothole on the road to board certification. As I look back, I don’t have enough fingers and toes to count all the good things that have happened to me, starting with Clem, my most influential mentor. Through hematology, I have had the opportunity to meet countless people brighter than me and to work with outstanding fellows and young scientists both here and abroad. And ASH became, and remains, an enormous part of my life. Hematology has been a marvelous journey, and I count myself most fortunate to have been able to make the trip. The Hematologist: ASH News and Reports EDITORS’ CHOICE ›› Thoughts From a Former Protégé F e b r u a r y 2 7, 2 0 1 4 Kenneth Kaushansky, MD Senior Vice President for Health Sciences, Dean School of Medicine, Stony Brook University In 1996, Hilary Clinton penned the book It Takes a Village to Raise a Child, based on an ancient African parable. I have often hijacked that title to claim, “It takes a village to raise a physician-scientist.” While I do believe that most successful physician-scientists can name multiple mentors, there is almost always, as John puts it, a “most important mentor.” Most scholarly articles on mentorship list at least six roles a successful mentor fills: teacher, sponsor, advisor, agent, role model, and confidante. Most would also agree that the successful mentor does not have the role of cloning oneself, but rather, he/she should be an astute enough listener to help the protégé pursue their own dreams – a seventh role. There is little doubt that John Adamson is my most important mentor, as he was able to fulfill each of these roles. It is no wonder that he was the 2013 recipient of the ASH Mentor Award. John taught me how to pen a scientific paper. When he revised and then handed back to me my very first scientific manuscript, I noticed it weighed about twice what it had when I handed it to him a few days earlier: the weight difference, of course, being that of the red ink. “Not bad for an author for whom English is a second language,” he quipped. But the weight of the red ink became less and less as time went on, as I learned not only science from John, but also how to communicate science. One of the critical junctures in my career came when I proudly pronounced that, despite not knowing which end of the pipette to use, I wanted to become a hematologic molecular biologist. John was a terrific sponsor; he made sure I had ample protected time for research, suggesting that purifying hematopoietic growth factors would help achieve my career goals and introducing me to another mentor, Earl Davie, who greatly enhanced my scientific toolbox. John has served as a primary advisor and academic confidante throughout my career, epitomizing the principle that mentorship does not end when the student leaves the lab. John was also a terrific agent. Shortly after we cloned human GM-CSF, John was invited to present his work on erythropoietin at a Gordon Research Conference (GRC). He suggested to the meeting organizers that I present in his place. It was my very first GRC. And John epitomizes the quintessential role model, constantly exuding scientific integrity, service to one’s community, and excellence in clinical care, educational endeavors, and scholarship. The Hematologist: ASH News and Reports JAK2. These observations suggest that CALR-mutated essential thrombocythemia is a distinct entity with a more indolent natural history. Dr. Bob Löwenberg (Editor-in-Chief) and Dr. Nancy Berliner (Deputy Editor-in-Chief) have combined efforts to identify some of the most outstanding Blood articles that have appeared either in print or online during the two-month interval between issues of The Hematologist. The citations are annotated to provide readers both with a concise description of the thrust of the article and an explanation of why the paper is particularly important. The goal is to underscore the remarkable research that is published in Blood and to highlight the exciting progress that is being made in the field. March 13, 2014 Sparkenbaugh EM, Chantrathammachart P, Mickelson J, et al. Differential contribution of FXa and thrombin to vascular inflammation in a mouse model of sickle cell disease. Blood. 2014;123:1747-1756. Both vascular inflammation and activation of coagulation contribute to the pathogenesis of sickle cell disease. In this study, Sparkenbaugh and colleagues used a mouse model of sickle cell disease (SCD) to demonstrate that inhibition of Factor Xa with rivaroxaban and inhibition of thrombin with dabigatran decrease lung neutrophil infiltration, and that rivaroxaban, but not dabigatran, decreases IL-6 levels. These observations could translate into development of novel therapies aimed at ameliorating the clinical manifestations of SCD. Hunter ZR, Xu L, Yang G, et al. The genomic landscape of Waldenstöm’s macroglobulinemia is characterized by highly recurring MYD88 and WHIM-like CXCR4 mutations, and small somatic deletions associated with B-cell lymphomagenesis. Blood. 2014;123:1637-1646. Hunter and colleagues report a major advance in our understanding of the genetic underpinnings of Waldenström’s macroglobulinemia (WM). They performed whole-genome sequencing on samples from 30 WM patients and subsequently validated findings of copy number alterations and somatic mutations in CXCR4 in expansion cohorts of 147 and 30 WM patients. The most striking findings were the identification of somatic mutations in MYD88 in 90 percent of patients (including the L256P activating mutation), in CXCR4 in 27 percent, and in ARID1A in 17 percent. Notably, the somatic CXCR4 mutations identified in this study included those observed in patients with the warts, hyogammaglobulinemia, infection and myelokathexis (WHIM) syndrome that had previously been reported only in the germ line. These findings, in addition to other recurring mutations identified in this study, contribute to a more complete understanding of the pathogenesis of WM. March 6, 2014 Rumi E, Pietra D, Ferretti V, et al. JAK2 or CALR mutation status defines subtypes of essential thrombocythemia with substantially different clinical course and outcomes. Blood. 2014;123:1544-1551. Rotunno G, Mannarelli C, Guglielmelli P, et al. Impact of calreticulin mutations on clinical and hematological phenotype and outcome in essential thrombocythemia. Blood. 2014;123:1552-1555. Recently, recurrent somatic calreticulin (CALR) insertion/ deletion mutations were discovered in the majority of patients with essential thrombocythemia and primary myelofibrosis in which neither JAK2 nor MPL was somatically mutated. In two independent studies, Rumi et al. and Rotunno et al. demonstrated that essential thrombocythemia patients with CALR mutations exhibit lower leukocyte and hemoglobin values, higher platelet counts, and a lower thrombosis risk as compared with patients with essential thrombocytosis with mutant Shim YK, Rachel JM, Ghia P, et al. Monoclonal B-cell lymphocytosis in healthy blood donors: an unexpectedly common finding. Blood. 2014;123:1319-1326. The prevalence of monoclonal B-cell lymphocytosis (MBL) in the general population ranges from <1 percent to 18 percent depending upon the detection method and population tested. Although a precursor to chronic lymphocytic leukemia, few people with MBL go on to develop clinically significant disease. In this Blood article, Shim and colleagues report the surprising observation that 7.1 percent of healthy blood donors have MBL. Most patients had “low-count” MBL (< 500 cells/L), but a small number had clinical MBL (> 500 cells/L). Although the risk of engraftment of these premalignant cells is low, the current findings raise questions about the need to screen for MBL in donated blood. At the very least, retrospective study of the outcomes of transfusion of MBL-containing units is warranted. Fe b r uary 20, 2014 Benveniste P, Serra P, Dervovic D, et al. Notch signals are required for in vitro but not in vivo maintenance of human hematopoietic stem cells and delay the appearance of multipotent progenitors. Blood. 2014;123:1167-1177. In murine model systems, Notch appears to be a pivotal signaling molecule that promotes the maintenance of “stemness” and expansion of self-renewing HSCs. And human CD34+ cells expand in vitro in response to Notch signals. In the current study, Benveniste and colleagues address whether Notch directly affects all hematopoietic stem cells (HSCs) and whether this role is relevant in vivo. The results clarify essential effects of Notch signaling on human HSC maintenance and expansion, and reconcile discrepancies in the literature about the role of Notch signaling in HSC homeostasis. Their findings support the concept that Notch signals maintain human HSCs in vitro that have hematopoietic-reconstituting capacity in vivo and delay the appearance of two newly described early progenitor cells. Fe b r uary 13, 2014 Aguilar R, Magallon-Tejada A, Achtman AH, et al. Molecular evidence for the localization of Plasmodium falciparum immature gametocytes in the bone marrow. Blood. 2014;123:959-966. Success in eradicating malaria hinges in part on the ability to prevent transmission of the parasite from the infected host back to the mosquito. In this article, Aguilar and colleagues reveal how complex an issue that may be. They present the first study based on quantitative polymerase chain reaction-based molecular analysis that delineates the characteristics of bone marrow and peripheral blood reservoirs of the parasite. They demonstrate that malarial sexual stages mature in the bone marrow and that hematologic manifestations (severe anemia and dyserythropoiesis) correlate with higher prevalence of mature gametocytes in the marrow. More important perhaps is the observation that mature, infectious gametocytes predominate in the peripheral blood, and that many asymptomatic anemic children harbor unexpectedly large numbers of circulating gametocytes. Such studies that define quantitatively and qualitatively the host-parasite relationship are a critical first step in developing strategies aimed at eradicating the malaria reservoir in humans. 15 wha t ’ s on t h e MARK YOUR C ALENDAR w e b As technology and the Web have evolved, so too have ASH’s online offerings. Now you can download ASH apps for your smartphone or tablet, follow ASH on Twitter (www.twitter.com/ ASH_hematology), find ASH videos on YouTube (www.youtube. com/user/ASHWebmaster), and visit ASH on Facebook at www.facebook.com/AmericanSocietyofHematology. ASH Seeks Volunteers for Website Users Group San Francisco, CA, December 6-9 May 1 Scholar Awards letter of intent due Washington, DC www.hematology.org/awards 2Application deadline for the ASH Visitor Training Program Washington, DC www.hematology.org/awards 14-17American Society of Pediatric Hematology/Oncology Annual Meeting Chicago, IL www.aspho.org 21-24 American Society of Gene & Cell Therapy Annual Meeting Washington, DC www.asgct.org 29-30Hematology for the Oncologist Seminar Chicago, IL www.asco.org 30-American Society of Clinical Oncology Annual Meeting June 3Chicago, IL As was mentioned in the cover story, a new site design was launched on April 4, as part of a multi-year effort to upgrade the Society’s technology platforms. ASH staff will be engaging in ongoing user testing to refine the website user experience. ASH members who are interested in being part of this process are encouraged to email webmaster@ hematology.org to sign up for the Website Users Group. Members of this informal group will be invited to act as beta testers and to provide feedback prior to the rollout of new website features. www.asco.org June 1 Application deadline for ASH Bridge Grant Washington, DC www.hematology.org/awards 5ASH annual meeting abstract submission site opens San Francisco, CA www.hematology.org/meetings 5Scholar Awards application available* Washington, DC www.hematology.org/awards 12-1519th Congress of European Hematology Association Milan, Italy www.ehaweb.org July 2Nomination deadline for the 2015 ASH Honorific Awards Washington, DC www.hematology.org/awards 24Members-only registration and housing opens for 2014 ASH Annual Meeting and Exposition San Francisco, CA www.hematology.org/meetings August 1 ASH Active and International Membership application deadline Washington, DC www.hematology.org/membership 1 Application deadline for Translational Research Training in Hematology Washington, DC www.hematology.org/awards 1 Application deadline for Scholar Awards Read The Hematologist online at Washington, DC www.hematology.org/awards www.hematology.org/thehematologist, 5Deadline to submit abstracts for the ASH annual meeting and catch up on the latest news in San Francisco, CA the field of hematology right on your 10-13ASH Meeting on Lymphoma Biology desktop, mobile phone, or tablet. Colorado Springs, CO www.hematology.org/meetings www.hematology.org/meetings *In order to submit an application for the Scholar Awards, you must have submitted a letter of intent by May 1, 2014. For additional meetings dates, go to www.hematology.org/Meetings/Non-ASH.aspx.