51
|
Truong TH, Pole JD, Bittencourt H, Schechter T, Cuvelier GD, Paulson K, Rayar M, Mitchell D, Schultz KR, O'Shea D, Barber R, Wall D, Sung L. Access to Hematopoietic Stem Cell Transplantation among Pediatric Patients with Acute Lymphoblastic Leukemia: A Population-Based Analysis. Biol Blood Marrow Transplant 2019; 25:1172-1178. [DOI: 10.1016/j.bbmt.2019.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 02/06/2019] [Indexed: 12/17/2022]
|
52
|
Abstract
PURPOSE OF REVIEW Incorporation of minimal residual disease (MRD) testing in acute lymphoblastic leukemia (ALL) and acute myeloblastic leukemia (AML) has transformed the landscape of hematopoietic cell transplantation (HCT). Pre-HCT MRD has allowed prognostication of HCT outcomes for high-risk leukemia patients, whereas the detection of post-HCT MRD has allowed for interventions to decrease relapse. RECENT FINDINGS In this review, we emphasize studies from the past two decades that highlight the critical role of MRD in HCT in pediatric ALL and AML. Advances in MRD detection methodology, using next-generation sequencing, have improved the sensitivity of MRD testing allowing for more accurate predictions of HCT outcomes for patients with relapsed and refractory ALL and AML. In addition, novel pre-HCT therapies, especially immunotherapy in ALL, have dramatically increased the number of patients who achieve MRD-negative remissions pre-HCT, resulting in improved HCT outcomes. Post-HCT MRD remains a challenge and new therapeutic interventions are needed to reduce post-HCT relapse. SUMMARY As immunotherapy increases pre-HCT MRD-negative remissions, and next-generation sequencing-MRD is incorporated to improve the sensitivity of MRD detection, future clinical studies will investigate less toxic HCT approaches to reduce long-term sequelae and to identify which patients may benefit most from early post-HCT intervention to reduce relapse.
Collapse
Affiliation(s)
- Agne Taraseviciute
- Division of Pediatric Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | | |
Collapse
|
53
|
Ifversen M, Turkiewicz D, Marquart HV, Winiarski J, Buechner J, Mellgren K, Arvidson J, Rascon J, Körgvee LT, Madsen HO, Abrahamsson J, Lund B, Jonsson OG, Heilmann C, Heyman M, Schmiegelow K, Vettenranta K. Low burden of minimal residual disease prior to transplantation in children with very high risk acute lymphoblastic leukaemia: The NOPHO ALL2008 experience. Br J Haematol 2019; 184:982-993. [DOI: 10.1111/bjh.15761] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/11/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Marianne Ifversen
- Department of Paediatric and Adolescent Medicine, Rigshospitalet; Copenhagen University Hospital; Copenhagen Denmark
| | | | - Hanne V. Marquart
- The Tissue Typing Laboratory; Department of Clinical Immunology; Copenhagen University Hospital, Rigshospitalet; Copenhagen Denmark
| | - Jacek Winiarski
- Astrid Lindgren Children's Hospital and Clintec; Karolinska University Hospital, Huddinge; Stockholm Sweden
| | - Jochen Buechner
- Department of Paediatric Haematology and Oncology; Oslo University Hospital; Oslo Norway
| | - Karin Mellgren
- Institution for Clinical Sciences; Department of Paediatrics; Queen Silvia Children's Hospital; Gothenburg Sweden
| | | | - Jelena Rascon
- Centre for Paediatric Oncology and Haematology; Children's Hospital; Vilnius University Hospital; Vilnius Lithuania
| | | | - Hans O. Madsen
- The Tissue Typing Laboratory; Department of Clinical Immunology; Copenhagen University Hospital, Rigshospitalet; Copenhagen Denmark
| | - Jonas Abrahamsson
- Institution for Clinical Sciences; Department of Paediatrics; Queen Silvia Children's Hospital; Gothenburg Sweden
| | - Bendik Lund
- Department of Paediatrics; St. Olavs University Hospital Trondheim; Trondheim Norway
- Department of Clinical and Molecular Medicine; NTNU; Trondheim Norway
| | | | - Carsten Heilmann
- Department of Paediatric and Adolescent Medicine, Rigshospitalet; Copenhagen University Hospital; Copenhagen Denmark
| | - Mats Heyman
- Childhood Cancer Research Unit; Karolinska Institute; Astrid Lindgren's Children's Hospital; Karolinska University Hospital; Stockholm Sweden
| | - Kjeld Schmiegelow
- Department of Paediatric and Adolescent Medicine, Rigshospitalet; Copenhagen University Hospital; Copenhagen Denmark
- Institute of Clinical Medicine; University of Copenhagen; Copenhagen Denmark
| | - Kim Vettenranta
- Department of Paediatrics; University of Helsinki; Helsinki Finland
| |
Collapse
|
54
|
Murine pre-B-cell ALL induces T-cell dysfunction not fully reversed by introduction of a chimeric antigen receptor. Blood 2018; 132:1899-1910. [PMID: 30209120 DOI: 10.1182/blood-2017-12-815548] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 08/25/2018] [Indexed: 12/13/2022] Open
Abstract
Adoptive transfer of patient-derived T cells modified to express chimeric antigen receptors (CARTs) has demonstrated dramatic success in relapsed/refractory pre-B-cell acute lymphoblastic leukemia (ALL), but response and durability of remission requires exponential CART expansion and persistence. Tumors are known to affect T-cell function, but this has not been well studied in ALL and in the context of chimeric antigen receptor (CAR) expression. Using TCF3/PBX1 and MLL-AF4-driven murine ALL models, we assessed the impact of progressive ALL on T-cell function in vivo. Vaccines protect against TCF3/PBX1.3 but were ineffective when administered after leukemia injection, suggesting immunosuppression induced early during ALL progression. T cells from leukemia-bearing mice exhibited increased expression of inhibitory receptors, including PD1, Tim3, and LAG3, and were dysfunctional following adoptive transfer in a model of T-cell receptor (TCR)-dependent leukemia clearance. Although expression of inhibitory receptors has been linked to TCR signaling, pre-B-cell ALL induced inhibitory receptor expression, at least in part, in a TCR-independent manner. Finally, introduction of a CAR into T cells generated from leukemia-bearing mice failed to fully reverse poor in vivo function.
Collapse
|
55
|
Methods and role of minimal residual disease after stem cell transplantation. Bone Marrow Transplant 2018; 54:681-690. [PMID: 30116018 DOI: 10.1038/s41409-018-0307-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/28/2018] [Accepted: 06/13/2018] [Indexed: 11/08/2022]
Abstract
Relapse is the major cause of treatment failure after stem cell transplantation. Despite the fact that relapses occurred even if transplantation was performed in complete remission, it is obvious that minimal residual disease is present though not morphologically evident. Since adaptive immunotherapy by donor lymphocyte infusion or other novel cell therapies as well as less toxic drugs, which can be used after transplantation, the detection of minimal residual disease (MRD) has become a clinical important variable for outcome. Besides the increasing options to treat MRD, the most advanced technologies currently allow to detect residual malignant cells with a sensitivity of 10-5 to 10-6.Under the patronage of the European Society for Blood and Marrow Transplantation (EBMT) and the American Society for Blood and Marrow Transplantation (ASBMT) the 3rd workshop was held on 4/5 November 2016 in Hamburg/Germany, with the aim to present an up-to-date status of epidemiology and biology of relapse and to summarize the currently available options to prevent and treat post-transplant relapse. Here the current methods and role of minimal residual disease for myeloid and lymphoid malignancies are summarized.
Collapse
|
56
|
Slayton WB, Schultz KR, Kairalla JA, Devidas M, Mi X, Pulsipher MA, Chang BH, Mullighan C, Iacobucci I, Silverman LB, Borowitz MJ, Carroll AJ, Heerema NA, Gastier-Foster JM, Wood BL, Mizrahy SL, Merchant T, Brown VI, Sieger L, Siegel MJ, Raetz EA, Winick NJ, Loh ML, Carroll WL, Hunger SP. Dasatinib Plus Intensive Chemotherapy in Children, Adolescents, and Young Adults With Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia: Results of Children's Oncology Group Trial AALL0622. J Clin Oncol 2018; 36:2306-2314. [PMID: 29812996 DOI: 10.1200/jco.2017.76.7228] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Addition of imatinib to intensive chemotherapy improved survival for children and young adults with Philadelphia chromosome-positive acute lymphoblastic leukemia. Compared with imatinib, dasatinib has increased potency, CNS penetration, and activity against imatinib-resistant clones. Patients and Methods Children's Oncology Group (COG) trial AALL0622 (Bristol Myers Squibb trial CA180-204) tested safety and feasibility of adding dasatinib to intensive chemotherapy starting at induction day 15 in patients with newly diagnosed Philadelphia chromosome-positive acute lymphoblastic leukemia age 1 to 30 years. Allogeneic hematopoietic stem-cell transplantation (HSCT) was recommended for patients at high risk based on slow response and for those with a matched family donor regardless of response after at least 11 weeks of therapy. Patients at standard risk based on rapid response received chemotherapy plus dasatinib for an additional 120 weeks. Patients with overt CNS leukemia received cranial irradiation. Results Sixty eligible patients were enrolled. Five-year overall (OS) and event-free survival rates (± standard deviations [SD]) were 86% ± 5% and 60% ± 7% overall, 87% ± 5% and 61% ± 7% for standard-risk patients (n = 48; 19% underwent HSCT), and 89% ± 13% and 67% ± 19% for high-risk patients (n = 9; 89% underwent HSCT), respectively. Five-year cumulative incidence (± SD) of CNS relapse was 15% ± 6%. Outcomes (± SDs) were similar to those in COG AALL0031, which used the same chemotherapy with continuous imatinib: 5-year OS of 81% ± 6% versus 86% ± 5% ( P = .63) and 5-year disease-free survival of 68% ± 7% versus 60% ± 7% ( P = 0.31) for AALL0031 versus AALL0622, respectively. IKZF1 deletions, present in 56% of tested patients, were associated with significantly inferior OS and event-free survival overall and in standard-risk patients. Conclusion Dasatinib was well tolerated with chemotherapy and provided outcomes similar to those with imatinib in COG AALL0031, where all patients received cranial irradiation. Our results support limiting HSCT to slow responders and suggest a potential role for transplantation in rapid responders with IKZF1 deletions.
Collapse
Affiliation(s)
- William B Slayton
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Kirk R Schultz
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - John A Kairalla
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Meenakshi Devidas
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Xinlei Mi
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Michael A Pulsipher
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Bill H Chang
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Charles Mullighan
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Ilaria Iacobucci
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Lewis B Silverman
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Michael J Borowitz
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Andrew J Carroll
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Nyla A Heerema
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Julie M Gastier-Foster
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Brent L Wood
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Sherri L Mizrahy
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Thomas Merchant
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Valerie I Brown
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Lance Sieger
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Marilyn J Siegel
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Elizabeth A Raetz
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Naomi J Winick
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Mignon L Loh
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - William L Carroll
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| | - Stephen P Hunger
- William B. Slayton, John A. Kairalla, Meenakshi Devidas, Xinlei Mi, and Sherri L. Mizrahy, University of Florida, Gainesville, FL; Kirk R. Schultz, BC Children's Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Michael A. Pulsipher, Children's Hospital of Los Angeles, Los Angeles; Lance Sieger, University of California Los Angles-Harbor, Torrance; Mignon L. Loh, University of California San Francisco, San Francisco, CA; Bill H. Chang, Oregon Health and Science University, Portland, OR; Charles Mullighan, Ilaria Iacobucci, and Thomas Merchant, St Jude's Research Hospital, Memphis, TN; Lewis B. Silverman, Dana-Farber Cancer Institute, Boston, MA; Michael J. Borowitz, Johns Hopkins University, Baltimore, MD; Andrew J. Carroll, University of Alabama at Birmingham, Birmingham, AL; Nyla A. Heerema, Ohio State University; Julie M. Gastier-Foster, Nationwide Children's Hospital, Columbus, OH; Brent L. Wood, University of Washington Seattle, Seattle, WA; Valerie I. Brown, Penn State Health Children's Hospital, Hershey; Stephen P. Hunger, Children's Hospital of Philadelphia, Philadelphia, PA; Marilyn J. Siegel, Washington University School of Medicine, St Louis, MO; Elizabeth A. Raetz, University of Utah, Salt Lake City, UT; Naomi J. Winick, University of Texas Southwestern Medical Center, Dallas, TX; and William L. Carroll, New York University Langone Health Center, New York, NY
| |
Collapse
|
57
|
Kato M, Kurata M, Kanda J, Kato K, Tomizawa D, Kudo K, Yoshida N, Watanabe K, Shimada H, Inagaki J, Koh K, Goto H, Kato K, Cho Y, Yuza Y, Ogawa A, Okada K, Inoue M, Hashii Y, Teshima T, Murata M, Atsuta Y. Impact of graft-versus-host disease on relapse and survival after allogeneic stem cell transplantation for pediatric leukemia. Bone Marrow Transplant 2018; 54:68-75. [PMID: 29795428 DOI: 10.1038/s41409-018-0221-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 04/16/2018] [Accepted: 04/22/2018] [Indexed: 11/09/2022]
Abstract
Graft-versus-host disease (GVHD) occasionally leads to morbidity and mortality but is thought to reduce the risk of relapses in patients with a hematological malignancy. However, information on the effect of GVHD in pediatric leukemia is limited. Using a nationwide registry, we retrospectively analyzed 1526 children who underwent allogeneic stem cell transplantation for leukemia. Grades 0-I acute GVHD were associated with a higher relapse rate at three years after transplantation, at 25.4 and 24.3%, respectively, than grades II, III, or IV acute GVHD at 18.9%, 21.2%, and 2.6%, respectively. In contrast, the overall survival curve of the grades 0 and I GVHD groups (79.0% and 79.5%, respectively) approximated that of the grade II GVHD group (76.3%), and the probability of survival was worst in the severe GVHD groups (66.9% for grade III and 42.5% for grade IV). Chronic GVHD also reduced the relapse risk but conferred no survival advantage. Acute lymphoblastic leukemia was more sensitive to acute GVHD than acute myeloid leukemia (AML) while AML was more sensitive to chronic GVHD. Our study reproduced the preventive effects of GVHD against pediatric leukemia relapses but failed to demonstrate a significant survival advantage.
Collapse
Affiliation(s)
- Motohiro Kato
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan. .,Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Tokyo, Japan.
| | - Mio Kurata
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagoya, Japan
| | - Junya Kanda
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koji Kato
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya, Japan
| | - Daisuke Tomizawa
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Kazuko Kudo
- Department of Pediatrics, Fujita Health University, Toyoake, Japan
| | - Nao Yoshida
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya, Japan
| | - Kenichiro Watanabe
- Department of Hematology and Oncology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Hiroyuki Shimada
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Jiro Inagaki
- Department of Pediatrics, National Kyushu Cancer Center, Fukuoka, Japan
| | - Katsuyoshi Koh
- Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Hiroaki Goto
- Division of Hemato-oncology/Regenerative Medicine, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Keisuke Kato
- Department of Hematology/Oncology, Ibaraki Children's Hospital, Mito, Japan
| | - Yuko Cho
- Department of Pediatrics, Hokkaido University Hospital, Sapporo, Japan
| | - Yuki Yuza
- Department of Hematology/Oncology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Atsushi Ogawa
- Department of Pediatrics, Niigata Cancer Center Hospital, Niigata, Japan
| | - Keiko Okada
- Department of Pediatric Hematology and Oncology, Osaka City General Hospital, Osaka, Japan
| | - Masami Inoue
- Department of Hematology/Oncology, Osaka Women's and Children's Hospital, Izumi, Japan
| | - Yoshiko Hashii
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takanori Teshima
- Department of Hematology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Makoto Murata
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiko Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagoya, Japan.,Department of Healthcare Administration, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
58
|
Hematopoietic Stem Cell Transplantation for Adult Philadelphia-Negative Acute Lymphoblastic Leukemia in the First Complete Remission in the Era of Minimal Residual Disease. Curr Oncol Rep 2018; 20:36. [PMID: 29577208 DOI: 10.1007/s11912-018-0679-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review is to discuss the potential role of allogeneic hematopoietic stem cell transplantation (allo-HSCT) for Philadelphia-negative (Ph-) adult acute lymphoblastic leukemia (ALL) in first complete remission (CR1) in the era of minimal residual disease (MRD). RECENT FINDINGS Allo-HSCT continues to have a role in the therapy of a selected group of high-risk adult patients with ALL in CR1. Although the clinical significance of MRD has been studied less extensively in adults with ALL than in children, recent studies support its role as the strongest prognostic factor that can identify patients that are unlikely to be cured by standard chemotherapy and benefit from undergoing allo-HSCT. In addition, MRD status both pre- and post-HSCT has been found to correlate directly with the risk of relapse. Currently, the clinical challenge consists on applying MRD and molecular failure to integrate novel agents and immunotherapy to lower MRD before allo-HSCT and to modulate the graft versus leukemia (GVL) effect after transplant.
Collapse
|
59
|
Elgarten CW, Arnold DE, Bunin NJ, Seif AE. Outcomes of matched sibling donor bone marrow transplantation in children using single-agent calcineurin inhibitors as prophylaxis for graft versus host disease. Pediatr Blood Cancer 2018; 65:10.1002/pbc.26726. [PMID: 28748621 PMCID: PMC5699955 DOI: 10.1002/pbc.26726] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/20/2017] [Accepted: 06/22/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Optimal graft versus host disease (GVHD) prophylaxis prevents severe manifestations without excess immunosuppression. Standard prophylaxis includes a calcineurin inhibitor (CNI) with low-dose methotrexate. However, single-agent CNI may be sufficient prophylaxis for a defined group of patients. Single-agent CNI has been used for GVHD prophylaxis for human leukocyte antigen (HLA)-matched sibling donor (MSD) bone marrow transplants (BMTs) in young patients at the Children's Hospital of Philadelphia for over 20 years. Here, we describe outcomes using this prophylactic strategy in a recent cohort. PROCEDURE We performed a single-institution chart review and retrospective analysis of consecutive children undergoing MSD BMT who received single-agent CNI for GVHD prophylaxis between January 2002 and December 2014. RESULTS Fifty-two children with a median age of 6.1 years (interquartile range [IQR] 2.5-8.3) and donor age of 6 years (IQR 3-10), with malignant and nonmalignant diseases (n = 35 and 17, respectively) were evaluated. Forty-three (82.6%) received oral prophylaxis with single-agent tacrolimus after initial intravenous therapy. Rates of GVHD were consistent with reported rates on dual prophylaxis: the overall incidence of grades 2-4 acute GVHD was 25.5%, grades 3-4 GVHD 9.8%, and chronic GVHD 10.4%. The cumulative incidence of relapse among children with malignancy was 20% at a median of 237 days (IQR 194-318) post-transplant. Two-year overall survival was 82.7% (95% confidence interval [CI]: 69.4-90.6%) and event-free survival was 78.9% (95% CI: 65.1-87.7%). No patient experienced graft failure. CONCLUSIONS Single-agent CNI is a safe, effective approach to GVHD prophylaxis in young patients undergoing HLA-identical sibling BMT. Additionally, single-agent oral tacrolimus is a reasonable alternative to cyclosporine in this population.
Collapse
Affiliation(s)
- Caitlin W. Elgarten
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania,Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Danielle E. Arnold
- Division of Immunology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Nancy J. Bunin
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alix E. Seif
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
60
|
|
61
|
Qayed M, Wang T, Hemmer MT, Spellman S, Arora M, Couriel D, Alousi A, Pidala J, Abdel-Azim H, Aljurf M, Ayas M, Bitan M, Cairo M, Choi SW, Dandoy C, Delgado D, Gale RP, Hale G, Frangoul H, Kamble RT, Kharfan-Dabaja M, Lehman L, Levine J, MacMillan M, Marks DI, Nishihori T, Olsson RF, Hematti P, Ringden O, Saad A, Satwani P, Savani BN, Schultz KR, Seo S, Shenoy S, Waller EK, Yu L, Horowitz MM, Horan J. Influence of Age on Acute and Chronic GVHD in Children Undergoing HLA-Identical Sibling Bone Marrow Transplantation for Acute Leukemia: Implications for Prophylaxis. Biol Blood Marrow Transplant 2017; 24:521-528. [PMID: 29155316 DOI: 10.1016/j.bbmt.2017.11.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 11/02/2017] [Indexed: 10/18/2022]
Abstract
Relapse remains the major cause of mortality after hematopoietic cell transplantation (HCT) for pediatric acute leukemia. Previous research has suggested that reducing the intensity of calcineurin inhibitor-based graft-versus-host disease (GVHD) prophylaxis may be an effective strategy for abrogating the risk of relapse in pediatric patients undergoing matched sibling donor (MSD) HCT. We reasoned that the benefits of this strategy could be maximized by selectively applying it to those patients least likely to develop GVHD. We conducted a study of risk factors for GVHD, to risk-stratify patients based on age. Patients age <18 years with leukemia who received myeloablative, T cell-replete MSD bone marrow transplantation and calcineurin inhibitor-based GVHD prophylaxis between 2000 and 2013 and were entered into the Center for International Blood and Marrow Transplant Research registry were included. The cumulative incidence of grade II-IV acute GVHD (aGVHD) was 19%, that of grade II-IV aGVHD 7%, and that of chronic GVHD (cGVHD) was 16%. Compared with age 13 to 18 years, age 2 to 12 years was associated with a lower risk of grade II-IV aGVHD (hazard ratio [HR], .42; 95% confidence interval [CI], .26 to .70; P = .0008), grade II-IV aGVHD (HR, .24; 95% CI, .10 to .56; P = .001), and cGVHD (HR, .32; 95% CI, .19 to .54; P < .001). Compared with 2000-2004, the risk of grade II-IV aGVHD was lower in children undergoing transplantation in 2005-2008 (HR, .36; 95% CI, .20 to .65; P = .0007) and in 2009-2013 (HR, .24; 95% CI. .11 to .53; P = .0004). Similarly, the risk of grade III-IV aGVHD was lower in children undergoing transplantation in 2005-2008 (HR, .23; 95% CI, .08 to .65; P = .0056) and 2009-2013 (HR, .16; 95% CI, .04 to .67; P = .0126) compared with those doing so in 2000-2004. We conclude that aGVHD rates have decreased significantly over time, and that children age 2 to 12 years are at very low risk for aGVHD and cGVHD. These results should be validated in an independent analysis, because these patients with high-risk malignancies may be good candidates for trials of reduced GVHD prophylaxis.
Collapse
Affiliation(s)
- Muna Qayed
- Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Atlanta, Georgia.
| | - Tao Wang
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Division of Biostatistics, Institute for Health and Society, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Michael T Hemmer
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Stephen Spellman
- Center for International Blood and Marrow Transplant Research, National Marrow Donor Program/Be the Match, Minneapolis, Minnesota
| | - Mukta Arora
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota Medical Center, Minneapolis, Minnesota
| | - Daniel Couriel
- Department of Internal Medicine, Division of Hematology and Hematologic Malignancies, Utah Blood and Marrow Transplant Program, Salt Lake City, Utah
| | - Amin Alousi
- Department of Stem Cell Transplantation, Division of Cancer Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Joseph Pidala
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Hisham Abdel-Azim
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California
| | - Mahmoud Aljurf
- Department of Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Mouhab Ayas
- Department of Pediatric Hematology/Oncology, King Faisal Specialist Hospital and Research Center, Ridayh, Saudi Arabia
| | - Menachem Bitan
- Department of Pediatric Hematology/Oncology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Mitchell Cairo
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, New York Medical College, Valhalla, New York
| | - Sung Won Choi
- Department of Pediatrics and Communicable Diseases, The University of Michigan, Ann Arbor, Michigan
| | - Christopher Dandoy
- , Department of Pediatrics, Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - David Delgado
- Department of Pediatrics, Indiana University Hospital, Indianapolis, Indiana
| | - Robert Peter Gale
- Hematology Research Centre, Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | - Gregory Hale
- Department of Hematology/Oncology, Johns Hopkins All Children's Hospital, St. Petersburg, Florida
| | - Haydar Frangoul
- Pediatric Hematology - Oncology, The Children's Hospital at TriStar Centennial and Sarah Cannon Research Institute, Nashville, Tennessee
| | - Rammurti T Kamble
- Division of Hematology and Oncology, Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Mohamed Kharfan-Dabaja
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Leslie Lehman
- Department of Pediatrics - Hematology Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - John Levine
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Margaret MacMillan
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota Medical Center, Minneapolis, Minnesota
| | - David I Marks
- Adult Bone Marrow Transplant, University Hospitals Bristol NHS Trust, Bristol, United Kingdom
| | - Taiga Nishihori
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Richard F Olsson
- Division of Therapeutic Immunology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Centre for Clinical Research Sormland, Uppsala University, Uppsala, Sweden
| | - Peiman Hematti
- Division of Hematology/Oncology/Bone Marrow Transplantation, Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, Wisconsin
| | - Olov Ringden
- Division of Therapeutic Immunology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ayman Saad
- Division of Hematology/Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Prakash Satwani
- Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Department of Pediatrics, Columbia University Medical Center, New York, New York
| | - Bipin N Savani
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kirk R Schultz
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplant, British Columbia's Children's Hospital, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Sachiko Seo
- National Cancer Research Center, East Hospital, Kashiwa, Chiba, Japan
| | - Shalini Shenoy
- Department of Pediatrics - Hematology Oncology, Washington University, St. Louis, Missouri
| | - Edmund K Waller
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Lolie Yu
- Division of Hematology/Oncology, Center for Cancer and Blood Disorders, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Mary M Horowitz
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - John Horan
- Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
62
|
Berry DA, Zhou S, Higley H, Mukundan L, Fu S, Reaman GH, Wood BL, Kelloff GJ, Jessup JM, Radich JP. Association of Minimal Residual Disease With Clinical Outcome in Pediatric and Adult Acute Lymphoblastic Leukemia: A Meta-analysis. JAMA Oncol 2017; 3:e170580. [PMID: 28494052 DOI: 10.1001/jamaoncol.2017.0580] [Citation(s) in RCA: 377] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Minimal residual disease (MRD) refers to the presence of disease in cases deemed to be in complete remission by conventional pathologic analysis. Assessing the association of MRD status following induction therapy in patients with acute lymphoblastic leukemia (ALL) with relapse and mortality may improve the efficiency of clinical trials and accelerate drug development. Objective To quantify the relationships between event-free survival (EFS) and overall survival (OS) with MRD status in pediatric and adult ALL using publications of clinical trials and other databases. Data Sources Clinical studies in ALL identified via searches of PubMed, MEDLINE, and clinicaltrials.gov. Study Selection Our search and study screening process adhered to the PRISMA Guidelines. Studies that addressed EFS or OS by MRD status in patients with ALL were included; reviews, abstracts, and studies with fewer than 30 patients or insufficient MRD description were excluded. Data Extraction and Synthesis Study sample size, patient age, follow-up time, timing of MRD assessment (postinduction or consolidation), MRD detection method, phenotype/genotype (B cell, T cell, Philadelphia chromosome), and EFS and OS. Searches of PubMed and MEDLINE identified 566 articles. A parallel search on clinicaltrials.gov found 67 closed trials and 62 open trials as of 2014. Merging results of 2 independent searches and applying exclusions gave 39 publications in 3 arms of patient populations (adult, pediatric, and mixed). We performed separate meta-analyses for each of these 3 subpopulations. Results The 39 publications comprised 13 637 patients: 16 adult studies (2076 patients), 20 pediatric (11 249 patients), and 3 mixed (312 patients). The EFS hazard ratio (HR) for achieving MRD negativity is 0.23 (95% Bayesian credible interval [BCI] 0.18-0.28) for pediatric patients and 0.28 (95% BCI, 0.24-0.33) for adults. The respective HRs in OS are 0.28 (95% BCI, 0.19-0.41) and 0.28 (95% BCI, 0.20-0.39). The effect was similar across all subgroups and covariates. Conclusions and Relevance The value of having achieved MRD negativity is substantial in both pediatric and adult patients with ALL. These results are consistent across therapies, methods of and times of MRD assessment, cutoff levels, and disease subtypes. Minimal residual disease status warrants consideration as an early measure of disease response for evaluating new therapies, improving the efficiency of clinical trials, accelerating drug development, and for regulatory approval. A caveat is that an accelerated approval of a particular new drug using an intermediate end point, such as MRD, would require confirmation using traditional efficacy end points.
Collapse
Affiliation(s)
- Donald A Berry
- Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston
| | - Shouhao Zhou
- Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston
| | | | | | - Shuangshuang Fu
- University of Texas Health Science Center at Houston, Houston
| | | | - Brent L Wood
- University of Washington School of Medicine, St Louis, Missouri
| | | | | | | |
Collapse
|
63
|
Rheingold SR, Tasian SK, Whitlock JA, Teachey DT, Borowitz MJ, Liu X, Minard CG, Fox E, Weigel BJ, Blaney SM. A phase 1 trial of temsirolimus and intensive re-induction chemotherapy for 2nd or greater relapse of acute lymphoblastic leukaemia: a Children's Oncology Group study (ADVL1114). Br J Haematol 2017; 177:467-474. [PMID: 28295182 DOI: 10.1111/bjh.14569] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/05/2016] [Indexed: 01/19/2023]
Abstract
The phosphatidylinositol 3-kinase (PI3K)/mammalian (or mechanistic) target of rapamycin (mTOR) signalling pathway is commonly dysregulated in acute lymphoblastic leukaemia (ALL). A phase 1 trial of the mTOR inhibitor temsirolimus in combination with UKALL R3 re-induction chemotherapy was conducted in children and adolescents with second or greater relapse of ALL. The initial temsirolimus dose level (DL1) was 10 mg/m2 weekly × 3 doses. Subsequent patient cohorts received temsirolimus 7·5 mg/m2 weekly × 3 doses (DL0) or, secondary to toxicity, 7·5 mg/m2 weekly × 2 doses (DL-1). Sixteen patients were enrolled, 15 were evaluable for toxicity. Dose-limiting toxicity (DLT) occurred at all three dose levels and included hypertriglyceridaemia, mucositis, ulceration, hypertension with reversible posterior leucoencephalopathy, elevated gamma-glutamyltransferase or alkaline phosphatase and sepsis. The addition of temsirolimus to UKALL R3 re-induction therapy resulted in excessive toxicity and was not tolerable in children with relapsed ALL. However, this regimen induced remission in seven of fifteen patients. Three patients had minimal residual disease levels <0·01%. Inhibition of PI3K signalling was detected in patients treated at all dose levels of temsirolimus, but inhibition at an early time point did not appear to correlate with clinical responses at the end of re-induction therapy.
Collapse
Affiliation(s)
- Susan R Rheingold
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah K Tasian
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - James A Whitlock
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, ON, Canada
| | - David T Teachey
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Michael J Borowitz
- Department of Pathology, Sidney Kimmel Cancer Center and Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Xiaowei Liu
- Children's Oncology Group Operations Center, Monrovia, CA, USA
| | - Charles G Minard
- Dan L. Duncan Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA.,TX Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Elizabeth Fox
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | | - Susan M Blaney
- Dan L. Duncan Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA.,TX Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
64
|
Chen Y, Huang XJ, Liu KY, Chen H, Chen YH, Zhang XH, Wang Y, Wang FR, Han W, Wang JZ, Yan CH, Xu LP. Association between C-reactive protein levels in the first 1-3 days post-transplant and allogeneic immune reactions. Biomark Med 2017; 11:117-124. [PMID: 28111964 DOI: 10.2217/bmm-2016-0244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
AIM The purpose of this study was to determine whether C-reactive protein (CRP) in the first 1-3 days post-transplant could predict allogeneic immune reactions, including engraftment syndrome or acute graft-versus-host disease (GVHD), in pediatric haploidentical stem cell transplantation. PATIENTS & METHODS The study population comprised 175 consecutive pediatric patients. Receiver operating characteristic analysis was performed to identify the cut-off CRP value. RESULTS The high-CRP group (≥20.1 mg/l) was associated with an increased occurrence of engraftment syndrome (hazard ratio [HR] = 2.046; p = 0.008), II-IV acute GVHD (HR = 2.203; p = 0.001) and severe GVHD (HR = 6.371; p = 0.004). CONCLUSION Our data suggest that higher CRP during the first 1-3 days post-transplant could be a predictor of allogeneic immune reactions.
Collapse
Affiliation(s)
- Yao Chen
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, 11 Xizhimen South Street, Beijing 100044, PR China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, 11 Xizhimen South Street, Beijing 100044, PR China
| | - Kai-Yan Liu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, 11 Xizhimen South Street, Beijing 100044, PR China
| | - Huan Chen
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, 11 Xizhimen South Street, Beijing 100044, PR China
| | - Yu-Hong Chen
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, 11 Xizhimen South Street, Beijing 100044, PR China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, 11 Xizhimen South Street, Beijing 100044, PR China
| | - Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, 11 Xizhimen South Street, Beijing 100044, PR China
| | - Feng-Rong Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, 11 Xizhimen South Street, Beijing 100044, PR China
| | - Wei Han
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, 11 Xizhimen South Street, Beijing 100044, PR China
| | - Jing-Zhi Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, 11 Xizhimen South Street, Beijing 100044, PR China
| | - Chen-Hua Yan
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, 11 Xizhimen South Street, Beijing 100044, PR China
| | - Lan-Ping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, 11 Xizhimen South Street, Beijing 100044, PR China
| |
Collapse
|
65
|
Pharmacokinetics, Pharmacodynamics, and Pharmacogenomics of Immunosuppressants in Allogeneic Hematopoietic Cell Transplantation: Part II. Clin Pharmacokinet 2016; 55:551-93. [PMID: 26620047 DOI: 10.1007/s40262-015-0340-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Part I of this article included a pertinent review of allogeneic hematopoietic cell transplantation (alloHCT), the role of postgraft immunosuppression in alloHCT, and the pharmacokinetics, pharmacodynamics, and pharmacogenomics of the calcineurin inhibitors and methotrexate. In this article (Part II), we review the pharmacokinetics, pharmacodynamics, and pharmacogenomics of mycophenolic acid (MPA), sirolimus, and the antithymocyte globulins (ATG). We then discuss target concentration intervention (TCI) of these postgraft immunosuppressants in alloHCT patients, with a focus on current evidence for TCI and on how TCI may improve clinical management in these patients. Currently, TCI using trough concentrations is conducted for sirolimus in alloHCT patients. Several studies demonstrate that MPA plasma exposure is associated with clinical outcomes, with an increasing number of alloHCT patients needing TCI of MPA. Compared with MPA, there are fewer pharmacokinetic/dynamic studies of rabbit ATG and horse ATG in alloHCT patients. Future pharmacokinetic/dynamic research of postgraft immunosuppressants should include '-omics'-based tools: pharmacogenomics may be used to gain an improved understanding of the covariates influencing pharmacokinetics as well as proteomics and metabolomics as novel methods to elucidate pharmacodynamic responses.
Collapse
|
66
|
Lutz M, Mielke S. New perspectives on the use of mTOR inhibitors in allogeneic haematopoietic stem cell transplantation and graft-versus-host disease. Br J Clin Pharmacol 2016; 82:1171-1179. [PMID: 27245261 PMCID: PMC5061796 DOI: 10.1111/bcp.13022] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/27/2016] [Accepted: 05/30/2016] [Indexed: 12/17/2022] Open
Abstract
Inhibition of the mechanistic target of rapamycin (mTOR) has been exploited largely both in solid tumour oncology and solid organ transplantation. More recently mTOR inhibitors such as sirolimus and everolimus have been introduced to the field of allogeneic haematopoietic stem cell transplantation where their unique combination of immunosuppressive purposes offering reduced nephrotoxicity and potential antimalignant effects reflect a unique drug profile that has led to their widespread use in both prophylaxis and therapy of graft-versus-host disease (GVHD). On the other hand haematological insufficiency, infectious complications as well as vasculopathies, have been frequently reported as limiting toxicities. Here, we review both the retrospective and prospective experience available to date and stress the need for prospective registration trials to reduce off label use and improve patient safety by optimizing dosing and enhancing pharmacovigilance. Furthermore, we speculate on the future role of mTOR inhibitors in allogeneic haematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Mathias Lutz
- Department of Medicine A, Münster University Medical Center, Münster, Germany
| | - Stephan Mielke
- Department of Internal Medicine II, Würzburg University Medical Center, Würzburg, Germany.
| |
Collapse
|
67
|
Törlén J, Ringdén O, Garming-Legert K, Ljungman P, Winiarski J, Remes K, Itälä-Remes M, Remberger M, Mattsson J. A prospective randomized trial comparing cyclosporine/methotrexate and tacrolimus/sirolimus as graft-versus-host disease prophylaxis after allogeneic hematopoietic stem cell transplantation. Haematologica 2016; 101:1417-1425. [PMID: 27662016 DOI: 10.3324/haematol.2016.149294] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/01/2016] [Indexed: 12/29/2022] Open
Abstract
Improvement of graft-versus-host disease prophylaxis remains an important goal in allogeneic hematopoietic stem cell transplantation. Based on reports of possibly preferential properties of sirolimus, we compared the standard regimen of cyclosporine and methotrexate (n=106) with a combination of tacrolimus and sirolimus (n=103) as graft-versus-host disease prophylaxis after allogeneic hematopoietic stem cell transplantation in a prospective, open, randomized trial. The hypothesis was that the tacrolimus/sirolimus regimen would lead to less acute graft-versus-host disease and reduced transplant-related mortality. There was no significant difference in the cumulative incidence of acute graft-versus-host disease of grades II-IV (41% vs. 51%; P=0.19) or grades III-IV (13% vs. 7%; P=0.09) between the groups. Time to neutrophil engraftment (18 days vs. 17 days; P=0.24) was similar, but time to platelet engraftment was longer in cyclosporine/methotrexate patients (14 vs. 12 days; P<0.01). No significant differences in incidence of oropharyngeal mucositis, time to full donor chimerism, or number of cytomegalovirus infections were seen between the two treatment arms, and transplant-related toxicities were equally distributed. Triglyceride (P=0.005) and cholesterol (P=0.009) levels were higher in tacrolimus/sirolimus patients. Transplant-related mortality (18% vs. 12%; P=0.40) and 5-year overall survival (72% vs. 71%; P=0.71) were similar. Five-year relapse-free survival in patients with malignant diagnoses was 65% in the cyclosporine/methotrexate group and 63% in the tacrolimus/sirolimus group (P=0.73). We conclude that tacrolimus/sirolimus remains a valid and safe alternative to cyclosporine/methotrexate as graft-versus-host disease prophylaxis after allogeneic hematopoietic stem cell transplantation, with comparable transplant-related outcomes. The trial was registered at clinicaltrials.gov identifier: 00993343.
Collapse
Affiliation(s)
- Johan Törlén
- Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden .,Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Olle Ringdén
- Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden
| | - Karin Garming-Legert
- Division of Oral and Maxillofacial Surgery, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Per Ljungman
- Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden.,Department of Hematology, Karolinska University Hospital and Division of Hematology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jacek Winiarski
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Kari Remes
- Department of Internal Medicine, Turku University Hospital, Finland.,Turku University, Finland
| | | | - Mats Remberger
- Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden.,Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Mattsson
- Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden.,Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
68
|
Eculizumab therapy in adults with allogeneic hematopoietic cell transplant-associated thrombotic microangiopathy. Bone Marrow Transplant 2016; 51:1241-4. [PMID: 27064689 DOI: 10.1038/bmt.2016.87] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
69
|
Nguyen HD, Chatterjee S, Haarberg KMK, Wu Y, Bastian D, Heinrichs J, Fu J, Daenthanasanmak A, Schutt S, Shrestha S, Liu C, Wang H, Chi H, Mehrotra S, Yu XZ. Metabolic reprogramming of alloantigen-activated T cells after hematopoietic cell transplantation. J Clin Invest 2016; 126:1337-52. [PMID: 26950421 DOI: 10.1172/jci82587] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 01/21/2016] [Indexed: 12/13/2022] Open
Abstract
Alloreactive donor T cells are the driving force in the induction of graft-versus-host disease (GVHD), yet little is known about T cell metabolism in response to alloantigens after hematopoietic cell transplantation (HCT). Here, we have demonstrated that donor T cells undergo metabolic reprograming after allogeneic HCT. Specifically, we employed a murine allogeneic BM transplant model and determined that T cells switch from fatty acid β-oxidation (FAO) and pyruvate oxidation via the tricarboxylic (TCA) cycle to aerobic glycolysis, thereby increasing dependence upon glutaminolysis and the pentose phosphate pathway. Glycolysis was required for optimal function of alloantigen-activated T cells and induction of GVHD, as inhibition of glycolysis by targeting mTORC1 or 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) ameliorated GVHD mortality and morbidity. Together, our results indicate that donor T cells use glycolysis as the predominant metabolic process after allogeneic HCT and suggest that glycolysis has potential as a therapeutic target for the control of GVHD.
Collapse
|
70
|
Goker H, Malkan UY, Demiroglu H, Buyukasik Y. Chimeric antigen receptor T cell treatment in hematologic malignancies. Transfus Apher Sci 2016; 54:35-40. [DOI: 10.1016/j.transci.2016.01.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
71
|
Armand P, Kim HT, Sainvil MM, Lange PB, Giardino AA, Bachanova V, Devine SM, Waller EK, Jagirdar N, Herrera AF, Cutler C, Ho VT, Koreth J, Alyea EP, McAfee SL, Soiffer RJ, Chen YB, Antin JH. The addition of sirolimus to the graft-versus-host disease prophylaxis regimen in reduced intensity allogeneic stem cell transplantation for lymphoma: a multicentre randomized trial. Br J Haematol 2016; 173:96-104. [PMID: 26729448 DOI: 10.1111/bjh.13931] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 11/18/2015] [Indexed: 11/28/2022]
Abstract
Inhibition of the mechanistic target of rapamycin (mTOR) pathway has clinical activity in lymphoma. The mTOR inhibitor sirolimus has been used in the prevention and treatment of graft-versus-host disease (GVHD) after allogeneic haematopoietic stem cell transplantation (HSCT). A retrospective study suggested that patients with lymphoma undergoing reduced intensity conditioning (RIC) HSCT who received sirolimus as part of their GVHD prophylaxis regimen had a lower rate of relapse. We therefore performed a multicentre randomized trial comparing tacrolimus, sirolimus and methotrexate to standard regimens in adult patients undergoing RIC HSCT for lymphoma in order to assess the possible benefit of sirolimus on HSCT outcome. 139 patients were randomized. There was no difference overall in 2-year overall survival, progression-free survival, relapse, non-relapse mortality or chronic GVHD. However, the sirolimus-containing arm had a significantly lower incidence of grade II-IV acute GVHD (9% vs. 25%, P = 0·015), which was more marked for unrelated donor grafts. In conclusion, the addition of sirolimus for GVHD prophylaxis in RIC HSCT is associated with no increased overall toxicity and a lower risk of acute GVHD, although it does not improve survival; this regimen is an acceptable option for GVHD prevention in RIC HSCT. This trial is registered at clinicaltrials.gov (NCT00928018).
Collapse
Affiliation(s)
- Philippe Armand
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Haesook T Kim
- Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Paulina B Lange
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Angela A Giardino
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Veronika Bachanova
- Department of Medical Oncology, University of Minnesota, Minneapolis, MN, USA
| | - Steven M Devine
- Department of Medicine, The Ohio State University Comprehensive Cancer Center Ohio State University, Columbus, OH, USA
| | - Edmund K Waller
- Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Neera Jagirdar
- Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | | | - Corey Cutler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Vincent T Ho
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - John Koreth
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Edwin P Alyea
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Steven L McAfee
- Bone Marrow Transplant Unit, Division of Hematology/Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Robert J Soiffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Yi-Bin Chen
- Bone Marrow Transplant Unit, Division of Hematology/Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Joseph H Antin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
72
|
Murine allogeneic CD19 CAR T cells harbor potent antileukemic activity but have the potential to mediate lethal GVHD. Blood 2015; 127:1361-70. [PMID: 26660684 DOI: 10.1182/blood-2015-08-664250] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 12/06/2015] [Indexed: 12/11/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) persisting or relapsing following bone marrow transplantation (BMT) has a dismal prognosis. Success with chimeric antigen receptor (CAR) T cells offers an opportunity to treat these patients with leukemia-redirected donor-derived T cells, which may be more functional than T cells derived from patients with leukemia but have the potential to mediate graft-versus-host disease (GVHD). We, together with others, have previously demonstrated tumor-specific T-cell dysfunction in the allogeneic environment. Here, we studied CAR T-cell function following BMT using an immunocompetent murine model of minor mismatched allogeneic transplantation followed by donor-derived CD19-CAR T cells. Allogeneic donor-derived CD19-CAR T cells eliminated residual ALL with equal potency to those administered after syngeneic BMT. Surprisingly, allogeneic CAR T cells mediated lethal acute GVHD with early mortality, which is atypical for this minor mismatch model. We demonstrated that both allogeneic and syngeneic CAR T cells show initial expansion as effector T cells, with a higher peak but rapid deletion of allogeneic CAR T cells. Interestingly, CAR-mediated acute GVHD was only seen in the presence of leukemia, suggesting CAR-target interactions induced GVHD. Indeed, serum interleukin (IL)-6 was elevated only in the presence of both leukemia and CAR T cells, and IL-6 neutralization ameliorated the severity of GVHD in a delayed donor lymphocyte infusion model. Finally, allogeneic CD4(+) CAR T cells were responsible for GVHD, which correlated with their ability to produce IL-6 upon CAR stimulation. Altogether, we demonstrate that donor-derived allogeneic CAR T cells are active but have the capacity to drive GVHD.
Collapse
|
73
|
Furlan SN, Watkins B, Tkachev V, Flynn R, Cooley S, Ramakrishnan S, Singh K, Giver C, Hamby K, Stempora L, Garrett A, Chen J, Betz KM, Ziegler CGK, Tharp GK, Bosinger SE, Promislow DEL, Miller JS, Waller EK, Blazar BR, Kean LS. Transcriptome analysis of GVHD reveals aurora kinase A as a targetable pathway for disease prevention. Sci Transl Med 2015; 7:315ra191. [PMID: 26606970 PMCID: PMC4876606 DOI: 10.1126/scitranslmed.aad3231] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Graft-versus-host disease (GVHD) is the most common complication of hematopoietic stem cell transplant (HCT). However, our understanding of the molecular pathways that cause this disease remains incomplete, leading to inadequate treatment strategies. To address this, we measured the gene expression profile of nonhuman primate (NHP) T cells during acute GVHD. Utilizing microarray technology, we measured the expression profiles of CD3(+) T cells from five cohorts: allogeneic transplant recipients receiving (i) no immunoprophylaxis (No Rx), (ii) sirolimus monotherapy (Siro), (iii) tacrolimus-methotrexate (Tac-Mtx), as well as (iv) autologous transplant recipients (Auto) and (v) healthy controls (HC). This comparison allowed us to identify transcriptomic signatures specific for alloreactive T cells and determine the impact of both mTOR (mechanistic target of rapamycin) and calcineurin inhibition on GVHD. We found that the transcriptional profile of unprophylaxed GVHD was characterized by significant perturbation of pathways regulating T cell proliferation, effector function, and cytokine synthesis. Within these pathways, we discovered potentially druggable targets not previously implicated in GVHD, prominently including aurora kinase A (AURKA). Utilizing a murine GVHD model, we demonstrated that pharmacologic inhibition of AURKA could improve survival. Moreover, we found enrichment of AURKA transcripts both in allo-proliferating T cells and in sorted T cells from patients with clinical GVHD. These data provide a comprehensive elucidation of the T cell transcriptome in primate acute GVHD and suggest that AURKA should be considered a target for preventing GVHD, which, given the many available AURKA inhibitors in clinical development, could be quickly deployed for the prevention of GVHD.
Collapse
Affiliation(s)
- Scott N Furlan
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, the University of Washington, and the Fred Hutchinson Cancer Research Center, Seattle WA 98101, USA
| | | | - Victor Tkachev
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, the University of Washington, and the Fred Hutchinson Cancer Research Center, Seattle WA 98101, USA
| | - Ryan Flynn
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA
| | - Sarah Cooley
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA
| | | | - Karnail Singh
- Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Cindy Giver
- Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kelly Hamby
- Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Linda Stempora
- Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | - Jingyang Chen
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, the University of Washington, and the Fred Hutchinson Cancer Research Center, Seattle WA 98101, USA
| | - Kayla M Betz
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, the University of Washington, and the Fred Hutchinson Cancer Research Center, Seattle WA 98101, USA
| | | | - Gregory K Tharp
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Steven E Bosinger
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Daniel E L Promislow
- Department of Pathology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Jeffrey S Miller
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA
| | | | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA
| | - Leslie S Kean
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, the University of Washington, and the Fred Hutchinson Cancer Research Center, Seattle WA 98101, USA.
| |
Collapse
|
74
|
Satwani P, Kahn J, Jin Z. Making strides and meeting challenges in pediatric allogeneic hematopoietic cell transplantation clinical trials in the United States: Past, present and future. Contemp Clin Trials 2015; 45:84-92. [DOI: 10.1016/j.cct.2015.06.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 06/08/2015] [Accepted: 06/15/2015] [Indexed: 12/19/2022]
|
75
|
Ho MSH, Mei SHJ, Stewart DJ. The Immunomodulatory and Therapeutic Effects of Mesenchymal Stromal Cells for Acute Lung Injury and Sepsis. J Cell Physiol 2015; 230:2606-17. [PMID: 25913273 DOI: 10.1002/jcp.25028] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 04/21/2015] [Indexed: 12/20/2022]
Abstract
It is increasingly recognized that immunomodulation represents an important mechanism underlying the benefits of many stem cell therapies, rather than the classical paradigm of transdifferentiation and cell replacement. In the former paradigm, the beneficial effects of cell therapy result from paracrine mechanism(s) and/or cell-cell interaction as opposed to direct engraftment and repair of diseased tissue and/or dysfunctional organs. Depending on the cell type used, components of the secretome, including microRNA (miRNA) and extracellular vesicles, may be able to either activate or suppress the immune system even without direct immune cell contact. Mesenchymal stromal cells (MSCs), also referred to as mesenchymal stem cells, are found not only in the bone marrow, but also in a wide variety of organs and tissues. In addition to any direct stem cell activities, MSCs were the first stem cells recognized to modulate immune response, and therefore they will be the focus of this review. Specifically, MSCs appear to be able to effectively attenuate acute and protracted inflammation via interactions with components of both innate and adaptive immune systems. To date, this capacity has been exploited in a large number of preclinical studies and MSC immunomodulatory therapy has been attempted with various degrees of success in a relatively large number of clinical trials. Here, we will explore the various mechanism employed by MSCs to effect immunosuppression as well as review the current status of its use to treat excessive inflammation in the context of acute lung injury (ALI) and sepsis in both preclinical and clinical settings.
Collapse
Affiliation(s)
- Mirabelle S H Ho
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario
| | - Shirley H J Mei
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario
| | - Duncan J Stewart
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario
| |
Collapse
|
76
|
Jacoby E, Chen A, Loeb DM, Gamper CJ, Zambidis E, Llosa NJ, Huo J, Cooke KR, Jones R, Fuchs E, Luznik L, Symons HJ. Single-Agent Post-Transplantation Cyclophosphamide as Graft-versus-Host Disease Prophylaxis after Human Leukocyte Antigen-Matched Related Bone Marrow Transplantation for Pediatric and Young Adult Patients with Hematologic Malignancies. Biol Blood Marrow Transplant 2015; 22:112-8. [PMID: 26343947 DOI: 10.1016/j.bbmt.2015.08.034] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 08/31/2015] [Indexed: 01/16/2023]
Abstract
High-dose cyclophosphamide given after HLA-matched related and unrelated allogeneic bone marrow transplantation (BMT) for patients with hematologic malignancies is effective single-agent graft-versus-host disease (GVHD) prophylaxis in adults. Data describing outcomes for pediatric and young adult patients have not been reported. Between the years 2007 and 2013, 29 pediatric and young adult patients ages ≤21 years of age treated at our institution for high-risk hematologic malignancies underwent myeloablative HLA-matched related T cell-replete BMT. Eleven patients received post-transplantation cyclophosphamide (PTCy) as single-agent GVHD prophylaxis and were followed prospectively. Eighteen patients received calcineurin inhibitor (CNI)-based standard GVHD prophylaxis and were studied retrospectively as a control group. No acute GVHD (aGVHD) developed in patients receiving PTCy, whereas patients receiving CNI-based GVHD prophylaxis had cumulative incidences of grades II to IV and grades III and IV aGVHD of 27% and 5%, respectively. No patients receiving PTCy developed chronic GHVD, compared to 1 in the control group. Two-year overall survival was similar between the 2 groups (54% PTCy versus 58% CNI-based prophylaxis), as was event-free survival (42% PTCy versus 47% CNI-based). The 5-year cumulative incidence of relapse was 58% for PTCy and 42% for CNI-based GVHD prophylaxis (P = .45). These results suggest that PTCy is a safe and efficacious method of GVHD prophylaxis after an HLA-matched related BMT in the pediatric and young adult population that affords patients to be off all post-transplantation immunosuppression on day +5.
Collapse
Affiliation(s)
- Elad Jacoby
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Pediatric Oncology Branch, National Institutes of Health, Bethesda, Maryland
| | - Allen Chen
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - David M Loeb
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christopher J Gamper
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elias Zambidis
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nicolas J Llosa
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jeffrey Huo
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kenneth R Cooke
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rick Jones
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ephraim Fuchs
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Leo Luznik
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Heather J Symons
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
77
|
Pulsipher MA, Langholz B, Wall DA, Schultz KR, Bunin N, Carroll W, Raetz E, Gardner S, Goyal RK, Gastier-Foster J, Borowitz M, Teachey D, Grupp SA. Risk factors and timing of relapse after allogeneic transplantation in pediatric ALL: for whom and when should interventions be tested? Bone Marrow Transplant 2015; 50:1173-9. [PMID: 25961775 PMCID: PMC4573663 DOI: 10.1038/bmt.2015.103] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 02/17/2015] [Accepted: 03/11/2015] [Indexed: 11/09/2022]
Abstract
We previously showed that minimal residual disease (MRD) detection pre-hematopoietic cell transplant (HCT) and acute GvHD (aGvHD) independently predicted risk of relapse in pediatric ALL. In this study we further define risk by assessing timing of relapse and the effects of leukemia risk category and post-HCT MRD. By multivariate analysis, pre-HCT MRD <0.1% and aGvHD by day +55 were associated with decreased relapse and improved event-free survival (EFS). Intermediate leukemia risk status predicted decreased relapse, and improved EFS and overall survival (OS). Patients with pre-HCT MRD ⩾0.1% who did not develop aGvHD compared with those with MRD <0.1% who did develop aGvHD had much worse survival (2 years EFS 18% vs 71%; P=0.001, 2 years OS 46 vs 74%; P=0.04). Patients with pre-HCT MRD <0.1% who did not experience aGvHD had higher rates of relapse than those who did develop aGvHD (40% vs 13%; P= 0.008). Post-HCT MRD led to a substantial increase in relapse risk (HR=4.5, P<0.01). Patients at high risk of relapse can be defined after transplant using leukemia risk category, presence of MRD pre or post HCT, and occurrence of aGvHD. An optimal window to initiate intervention to prevent relapse occurs between day +55 and +200 after HCT.
Collapse
Affiliation(s)
- Michael A. Pulsipher
- Division of Hematology and Hematological Malignancies, Huntsman Cancer Institute/University of Utah School of Medicine, Primary Children’s Hospital, Salt Lake City, UT
| | - Bryan Langholz
- Department of Preventive Medicine, USC Keck School of Medicine, Los Angeles, CA
| | - Donna A. Wall
- Manitoba Blood and Marrow Transplant Program, Winnepeg, MB, Canada
| | - Kirk R. Schultz
- Department of Pediatrics University of BC, BC Children’s Hospital, Vancouver, BC, Canada
| | - Nancy Bunin
- Division of Oncology, Children’s Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - William Carroll
- NYU Department of Pediatrics and Cancer Institute, NYU Langone Medical Center, New York, NY
| | - Elizabeth Raetz
- Division of Hematology and Hematological Malignancies, Huntsman Cancer Institute/University of Utah School of Medicine, Primary Children’s Hospital, Salt Lake City, UT
| | - Sharon Gardner
- NYU Department of Pediatrics and Cancer Institute, NYU Langone Medical Center, New York, NY
| | - Rakesh K. Goyal
- Division of Blood and Marrow Transplantation and Cellular Therapies, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Julie Gastier-Foster
- Department of Pathology and Laboratory Medicine, Nationwide Children’s Hospital, Columbus, OH and Departments of Pathology and Pediatrics, The Ohio State University College of Medicine, Columbus, OH
| | - Michael Borowitz
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - David Teachey
- Division of Oncology, Children’s Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Stephan A. Grupp
- Division of Oncology, Children’s Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Pathology, Children’s Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
78
|
Burke MJ, Verneris MR, Le Rademacher J, He W, Abdel-Azim H, Abraham AA, Auletta JJ, Ayas M, Brown VI, Cairo MS, Chan KW, Diaz Perez MA, Dvorak CC, Egeler RM, Eldjerou L, Frangoul H, Guilcher GMT, Hayashi RJ, Ibrahim A, Kasow KA, Leung WH, Olsson RF, Pulsipher MA, Shah N, Shah NN, Thiel E, Talano JA, Kitko CL. Transplant Outcomes for Children with T Cell Acute Lymphoblastic Leukemia in Second Remission: A Report from the Center for International Blood and Marrow Transplant Research. Biol Blood Marrow Transplant 2015; 21:2154-2159. [PMID: 26327632 DOI: 10.1016/j.bbmt.2015.08.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 08/20/2015] [Indexed: 10/23/2022]
Abstract
Survival for children with relapsed T cell acute lymphoblastic leukemia (T-ALL) is poor when treated with chemotherapy alone, and outcomes after allogeneic hematopoietic cell transplantation (HCT) is not well described. Two hundred twenty-nine children with T-ALL in second complete remission (CR2) received an HCT after myeloablative conditioning between 2000 and 2011 and were reported to the Center for International Blood and Marrow Transplant Research. Median age was 10 years (range, 2 to 18). Donor source was umbilical cord blood (26%), matched sibling bone marrow (38%), or unrelated bone marrow/peripheral blood (36%). Acute (grades II to IV) and chronic graft-versus-host disease occurred in, respectively, 35% (95% confidence interval [CI], 27% to 45%) and 26% (95% CI, 20% to 33%) of patients. Transplant-related mortality at day 100 and 3-year relapse rates were 13% (95% CI, 9% to 18%) and 30% (95% CI, 24% to 37%), respectively. Three-year overall survival and disease-free survival rates were 48% (95% CI, 41% to 55%) and 46% (95% CI, 39% to 52%), respectively. In multivariate analysis, patients with bone marrow relapse, with or without concurrent extramedullary relapse before HCT, were most likely to relapse (hazard ratio, 3.94; P = .005) as compared with isolated extramedullary disease. In conclusion, HCT for pediatric T-ALL in CR2 demonstrates reasonable and durable outcomes, and consideration for HCT is warranted.
Collapse
Affiliation(s)
- Michael J Burke
- Division of Hematology/Oncology/Blood and Marrow Transplant, Department of Pediatrics, Medical College of Wisconsin and Children's Hospital of Wisconsin, Milwaukee, WI.
| | | | - Jennifer Le Rademacher
- CIBMTR(®) (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, WI; Division of Biostatistics, Institute for Health and Society, Medical College of Wisconsin, Milwaukee, WI
| | - Wensheng He
- CIBMTR(®) (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Hisham Abdel-Azim
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Allistair A Abraham
- Division of Blood and Marrow Transplantation, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC
| | - Jeffery J Auletta
- Divisions of Hematology/Oncology, Bone Marrow Transplantation and Infectious Diseases, Nationwide Children's Hospital, Columbus, OH
| | - Mouhab Ayas
- Department of Pediatric Hematology Oncology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Valerie I Brown
- Division of Pediatric Oncology/Hematology, Department of Pediatrics, Penn State Hershey Children's Hospital and College of Medicine, Hershey, PA
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY
| | - Ka Wah Chan
- Department of Pediatrics, Texas Transplant Institute, San Antonio, TX
| | - Miguel A Diaz Perez
- Department of Hematology/Oncology, Hospital Infantil Universitario Nino Jesus, Madrid, Spain
| | - Christopher C Dvorak
- Department of Pediatrics, University of California San Francisco Medical Center, San Francisco, CA
| | - R Maarten Egeler
- Department of Hematology/Oncology, Hospital for Sick Children, Toronto, ON, Canada
| | - Lamis Eldjerou
- Department of Pediatrics, University of Florida, Gainsville, FL
| | - Haydar Frangoul
- Division of Hematology-Oncology, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN
| | - Gregory M T Guilcher
- Section of Paediatric Oncology and Blood and Marrow Transplant, Alberta Children's Hospital, Calgary, AB, Canada
| | - Robert J Hayashi
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Ahmed Ibrahim
- Department of Hematology/Oncology, Makassed General Hospital, Beiruit, Lebanon
| | - Kimberly A Kasow
- Division of Hematology-Oncology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Wing H Leung
- Division of Bone Marrow Transplantation, St. Jude Children's Research Hospital, Memphis, TN
| | - Richard F Olsson
- Division of Therapeutic Immunology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Centre for Clinical Research Sörmland, Uppsala University, Uppsala, Sweden
| | - Michael A Pulsipher
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Niketa Shah
- Division of Hematology/Oncology, Department of Pediatrics, Mayo Clinic Arizona and Phoenix Children's Hospital, Phoenix, AZ
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NIH), Bethesda, MD
| | - Elizabeth Thiel
- CIBMTR(®) (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Julie-An Talano
- Division of Hematology/Oncology/Blood and Marrow Transplant, Department of Pediatrics, Medical College of Wisconsin and Children's Hospital of Wisconsin, Milwaukee, WI
| | - Carrie L Kitko
- Stem Cell Transplant Program, Department of Pediatrics, Vanderbilt University, Nashville, TN
| |
Collapse
|
79
|
Maude SL, Teachey DT, Porter DL, Grupp SA. CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood 2015; 125:4017-23. [PMID: 25999455 PMCID: PMC4481592 DOI: 10.1182/blood-2014-12-580068] [Citation(s) in RCA: 519] [Impact Index Per Article: 51.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 01/14/2015] [Indexed: 12/11/2022] Open
Abstract
Relapsed and refractory acute lymphoblastic leukemia (ALL) remains difficult to treat, with minimal improvement in outcomes seen in more than 2 decades despite advances in upfront therapy and improved survival for de novo ALL. Adoptive transfer of T cells engineered to express a chimeric antigen receptor (CAR) has emerged as a powerful targeted immunotherapy, showing striking responses in highly refractory populations. Complete remission (CR) rates as high as 90% have been reported in children and adults with relapsed and refractory ALL treated with CAR-modified T cells targeting the B-cell-specific antigen CD19. Distinct CAR designs across several studies have produced similar promising CR rates, an encouraging finding. Even more encouraging are durable remissions observed in some patients without additional therapy. Duration of remission and CAR-modified T-cell persistence require further study and more mature follow-up, but emerging data suggest these factors may distinguish CAR designs. Supraphysiologic T-cell proliferation, a hallmark of this therapy, contributes to both efficacy and the most notable toxicity, cytokine release syndrome (CRS), posing a unique challenge for toxicity management. This review will discuss the current landscape of CD19 CAR clinical trials, CRS pathophysiology and management, and remaining challenges.
Collapse
Affiliation(s)
- Shannon L Maude
- Division of Oncology, The Children's Hospital of Philadelphia, Department of Pediatrics
| | - David T Teachey
- Division of Oncology, The Children's Hospital of Philadelphia, Department of Pediatrics
| | | | - Stephan A Grupp
- Division of Oncology, The Children's Hospital of Philadelphia, Department of Pediatrics, Department of Pathology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
80
|
Pulsipher MA, Carlson C, Langholz B, Wall DA, Schultz KR, Bunin N, Kirsch I, Gastier-Foster JM, Borowitz M, Desmarais C, Williamson D, Kalos M, Grupp SA. IgH-V(D)J NGS-MRD measurement pre- and early post-allotransplant defines very low- and very high-risk ALL patients. Blood 2015; 125:3501-8. [PMID: 25862561 PMCID: PMC4447864 DOI: 10.1182/blood-2014-12-615757] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 03/26/2015] [Indexed: 01/10/2023] Open
Abstract
Positive detection of minimal residual disease (MRD) by multichannel flow cytometry (MFC) prior to hematopoietic cell transplantation (HCT) of patients with acute lymphoblastic leukemia (ALL) identifies patients at high risk for relapse, but many pre-HCT MFC-MRD negative patients also relapse, and the predictive power MFC-MRD early post-HCT is poor. To test whether the increased sensitivity of next-generation sequencing (NGS)-MRD better identifies pre- and post-HCT relapse risk, we performed immunoglobulin heavy chain (IgH) variable, diversity, and joining (V[D]J) DNA sequences J NGS-MRD on 56 patients with B-cell ALL enrolled in Children's Oncology Group trial ASCT0431. NGS-MRD predicted relapse and survival more accurately than MFC-MRD (P < .0001), especially in the MRD negative cohort (relapse, 0% vs 16%; P = .02; 2-year overall survival, 96% vs 77%; P = .003). Post-HCT NGS-MRD detection was better at predicting relapse than MFC-MRD (P < .0001), especially early after HCT (day 30 MFC-MRD positive relapse rate, 35%; NGS-MRD positive relapse rate, 67%; P = .004). Any post-HCT NGS positivity resulted in an increase in relapse risk by multivariate analysis (hazard ratio, 7.7; P = .05). Absence of detectable IgH-V(D)J NGS-MRD pre-HCT defines good-risk patients potentially eligible for less intense treatment approaches. Post-HCT NGS-MRD is highly predictive of relapse and survival, suggesting a role for this technique in defining patients early who would be eligible for post-HCT interventions. The trial was registered at www.clinicaltrials.gov as #NCT00382109.
Collapse
Affiliation(s)
- Michael A Pulsipher
- Division of Hematology and Hematological Malignancies, Huntsman Cancer Institute/University of Utah School of Medicine, Primary Children's Hospital, Salt Lake City, UT
| | - Chris Carlson
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA; Adaptive Biotechnologies, Seattle, WA
| | - Bryan Langholz
- Department of Preventive Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Donna A Wall
- Manitoba Blood and Marrow Transplant Program, Winnipeg, MB, Canada
| | - Kirk R Schultz
- Department of Pediatrics, University of British Columbia, BC Children's Hospital, Vancouver, BC, Canada
| | - Nancy Bunin
- Division of Oncology, Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - Julie M Gastier-Foster
- Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH; Department of Pathology and Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH
| | - Michael Borowitz
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
| | | | | | - Michael Kalos
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; and
| | - Stephan A Grupp
- Division of Oncology, Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Department of Pathology, Children's Hospital of Philadelphia, and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
81
|
Wang L, Gu Z, Zhai R, Li D, Zhao S, Luo L, Zhao X, Wei H, Pang Z, Wang L, Liu D, Wang Q, Gao C. The efficacy and safety of sirolimus-based graft-versus-host disease prophylaxis in patients undergoing allogeneic hematopoietic stem cell transplantation: a meta-analysis of randomized controlled trials. Transfusion 2015; 55:2134-41. [PMID: 25857725 DOI: 10.1111/trf.13110] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 01/16/2015] [Accepted: 03/03/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND The efficacy and safety of sirolimus (SIR)-based graft-versus-host disease (GVHD) prophylaxis in patients who were subjected to allogeneic hematopoietic stem cell transplantation (allo-HSCT) remain to be clarified; this meta-analysis was conducted to evaluate these factors. STUDY DESIGN AND METHODS Data from original research were obtained from PubMed, Embase, and Cochrane central register of controlled trials databases. Randomized controlled trials (RCTs) evaluating the efficacy of SIR-based prophylaxis in allo-HSCT were included. The risk ratio (RR), with a 95% confidence interval (CI), was used to pool data. The random effects model was used, irrespective of the presence or absence of heterogeneity. RESULTS Five RCTs were included in the meta-analysis. SIR was observed to significantly decrease the incidence of Grade II to IV acute GVHD (aGVHD; RR, 0.65; 95% CI, 0.47-0.89). However, the incidence of Grade III to IV aGVHD and chronic GVHD was not decreased (RR, 0.91; 95% CI, 0.59-1.40; RR, 1.04; 95% CI, 0.88-1.23, respectively). An analysis of the toxic effects of SIR revealed that SIR effected a significant increase in the incidence of sinusoidal obstructive syndrome (RR, 2.24; 95% CI, 1.26-4.01), while that of thrombotic microangiopathy was not significantly increased (RR, 2.48; 95% CI, 0.87-7.06). Moreover, SIR did not improve event-free survival and overall survival (RR, 0.97; 95% CI, 0.85-1.10; and RR, 0.92; 95% CI, 0.82-1.02, respectively). CONCLUSION This meta-analysis indicated that the SIR-based regimen is an effective and safe alternative prophylaxis strategy for GVHD.
Collapse
Affiliation(s)
- Li Wang
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhenyang Gu
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Ruiren Zhai
- Department of Hematology and Oncology, Laoshan Branch of No. 401 Hospital of Chinese PLA, Qingdao, China
| | - Dandan Li
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Shasha Zhao
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Lan Luo
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Xiaoli Zhao
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Huaping Wei
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhaoxia Pang
- Department of Hematology and Oncology, Laoshan Branch of No. 401 Hospital of Chinese PLA, Qingdao, China
| | - Lili Wang
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Daihong Liu
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Quanshun Wang
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Chunji Gao
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
82
|
Abstract
Together, acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML) make up approximately one-third of all pediatric cancer diagnoses. Despite remarkable improvement in the treatment outcomes of these diseases over the past several decades, the prognosis for certain high-risk groups of leukemia and for relapsed disease remains poor. However, recent insights into different types of 'driver' lesions of leukemogenesis, such as the aberrant activation of signaling pathways and various epigenetic modifications, have led to the discovery of novel agents that specifically target the mechanism of transformation. In parallel, emerging approaches in cancer immunotherapy have led to newer therapies that can exploit and harness cytotoxic immunity directed against malignant cells. This review details the rationale and implementation of recent and specifically targeted therapies in acute pediatric leukemia. Topics covered include the inhibition of critical cell signaling pathways [BCR-ABL, FMS-like tyrosine kinase 3 (FLT3), mammalian target of rapamycin (mTOR), and Janus-associated kinase (JAK)], proteasome inhibition, inhibition of epigenetic regulators of gene expression [DNA methyltransferase (DNMT) inhibitors, histone deacetylase (HDAC) inhibitors, and disruptor of telomeric signaling-1 (DOT1L) inhibitors], monoclonal antibodies and immunoconjugated toxins, bispecific T-cell engaging (BiTE) antibodies, and chimeric antigen receptor-modified (CAR) T cells.
Collapse
Affiliation(s)
- Colleen E Annesley
- Oncology and Pediatrics, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Patrick Brown
- Oncology and Pediatrics, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB-I 2M46, Baltimore, MD 21231, USA
| |
Collapse
|
83
|
Mazaira GI, Camisay MF, De Leo S, Erlejman AG, Galigniana MD. Biological relevance of Hsp90-binding immunophilins in cancer development and treatment. Int J Cancer 2015; 138:797-808. [PMID: 25754838 DOI: 10.1002/ijc.29509] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/17/2015] [Indexed: 12/14/2022]
Abstract
Immunophilins are a family of intracellular receptors for immunosuppressive drugs. Those immunophilins that are related to immunosuppression are the smallest proteins of the family, i.e., FKBP12 and CyPA, whereas the other members of the family have higher molecular weight because the show additional domains to the drug-binding site. Among these extra domains, the TPR-domain is perhaps the most relevant because it permits the interaction of high molecular weight immunophilins with the 90-kDa heat-shock protein, Hsp90. This essential molecular chaperone regulates the biological function of several protein-kinases, oncogenes, protein phosphatases, transcription factors and cofactors . Hsp90-binding immunophilins where first characterized due to their association with steroid receptors. They regulate the cytoplasmic transport and the subcellular localization of these and other Hsp90 client proteins, as well as transcriptional activity, cell proliferation, cell differentiation and apoptosis. Hsp90-binding immunophilins are frequently overexpressed in several types of cancers and play a key role in cell survival. In this article we analyze the most important biological actions of the best characterized Hsp90-binding immunophilins in both steroid receptor function and cancer development and discuss the potential use of these immunophilins for therapeutic purposes as potential targets of specific small molecules.
Collapse
Affiliation(s)
- Gisela I Mazaira
- Departamento De Química Biológica, Facultad De Ciencias Exactas Y Naturales, Universidad De Buenos Aires and IQUIBICEN-CONICET, Buenos Aires, Argentina
| | - María F Camisay
- Departamento De Química Biológica, Facultad De Ciencias Exactas Y Naturales, Universidad De Buenos Aires and IQUIBICEN-CONICET, Buenos Aires, Argentina
| | - Sonia De Leo
- Departamento De Química Biológica, Facultad De Ciencias Exactas Y Naturales, Universidad De Buenos Aires and IQUIBICEN-CONICET, Buenos Aires, Argentina
| | - Alejandra G Erlejman
- Departamento De Química Biológica, Facultad De Ciencias Exactas Y Naturales, Universidad De Buenos Aires and IQUIBICEN-CONICET, Buenos Aires, Argentina
| | - Mario D Galigniana
- Departamento De Química Biológica, Facultad De Ciencias Exactas Y Naturales, Universidad De Buenos Aires and IQUIBICEN-CONICET, Buenos Aires, Argentina.,Instituto De Biología Y Medicina Experimental-CONICET, Buenos Aires, Argentina
| |
Collapse
|
84
|
Appelbaum FR, Anasetti C, Antin JH, Atkins H, Davies S, Devine S, Giralt S, Heslop H, Laport G, Lee SJ, Logan B, Pasquini M, Pulsipher M, Stadtmauer E, Wingard JR, Horowitz MM. Blood and marrow transplant clinical trials network state of the Science Symposium 2014. Biol Blood Marrow Transplant 2015; 21:202-24. [PMID: 25445636 PMCID: PMC4426907 DOI: 10.1016/j.bbmt.2014.10.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 10/06/2014] [Indexed: 12/31/2022]
Affiliation(s)
- Frederick R Appelbaum
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.
| | - Claudio Anasetti
- Research & Clinical Trials, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Joseph H Antin
- Stem Cell Transplants, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Harold Atkins
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Stella Davies
- Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Steven Devine
- Blood and Marrow Transplant Program, The Ohio State University, Columbus, Ohio
| | - Sergio Giralt
- Bone Marrow Transplant Service, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Helen Heslop
- Adult Bone Marrow and Stem Cell Transplant Program, Baylor College of Medicine, Houston, Texas
| | - Ginna Laport
- Medicine-Blood & Marrow Transplantation, Stanford Hospital and Clinics, Stanford, California
| | - Stephanie J Lee
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Brent Logan
- Clinical Research Division, Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Marcelo Pasquini
- Clinical Research Division, Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Michael Pulsipher
- Biostatistics, University of Utah School of Medicine, Primary Children's Hospital, Salt Lake City, Utah
| | - Edward Stadtmauer
- Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John R Wingard
- Hematology Division-Internal Medicine Department, University of Florida, Gainesville, Florida
| | - Mary M Horowitz
- Clinical Research Division, Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
85
|
Horn B, Petrovic A, Wahlstrom J, Dvorak CC, Kong D, Hwang J, Expose-Spencer J, Gates M, Cowan MJ. Chimerism-based pre-emptive immunotherapy with fast withdrawal of immunosuppression and donor lymphocyte infusions after allogeneic stem cell transplantation for pediatric hematologic malignancies. Biol Blood Marrow Transplant 2015; 21:729-37. [PMID: 25644958 DOI: 10.1016/j.bbmt.2014.12.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 12/23/2014] [Indexed: 11/25/2022]
Abstract
The presence of increasing host chimerism or persistent mixed chimerism (MC) after hematopoietic stem cell transplantation for leukemia in children is a predictor of relapse. To reduce the risk of relapse, we prospectively studied post-transplantation chimerism-based immunotherapy (IT) using fast withdrawal of immunosuppression (FWI) and donor lymphocyte infusions (DLI) in children with early post-transplantation MC. Forty-three children with hematologic malignancies at 2 institutions were enrolled prospectively in this study from 2009 until 2012 and were followed for a mean of 42 (SD, 10) months. Twelve patients (28%) were assigned to the observation arm based on the presence of graft-versus-host disease (GVHD) or full donor chimerism (FDC), and 5 (12%) sustained early events and could not undergo intervention. Twenty-six (60%) patients with MC were assigned to IT with FWI, which started at a median of 49 days (range, 35 to 85 days) after transplantation. Fourteen patients proceeded to DLI after FWI. Toxicities of treatment included GVHD, which developed in 19% of patients undergoing intervention, with 1 of 26 (4%) dying from GVHD and 1 (4%) still requiring therapy for chronic GVHD 21 months after DLI. Patients with MC undergoing IT had similar 2-year event-free survival (EFS) (73%; 95% confidence interval (CI), 55% to 91%) compared with patients who achieved FDC spontaneously (83%; 95% CI, 62% to 100%); however, because 50% of all relapses in the IT occurred later than 2 years after transplantation, the EFS declined to 55% (95% CI, 34% to 76%) at 42 (SD, 11) months. There were no late relapses in the observation group. EFS in the entire cohort was 58% (95% CI, 42% to 73%) at 42 (SD, 11) months after transplantation. Evidence of disease before transplantation remained a significant predictor of relapse, whereas development of chronic GVHD was protective against relapse.
Collapse
Affiliation(s)
- Biljana Horn
- Allergy Immunology and Blood and Marrow Transplant Division at Benioff Children's Hospital at University of California San Francisco, San Francisco, California.
| | - Aleksandra Petrovic
- Department of Blood and Marrow Transplantation, All Children's Hospital, St. Petersburg, Florida
| | - Justin Wahlstrom
- Allergy Immunology and Blood and Marrow Transplant Division at Benioff Children's Hospital at University of California San Francisco, San Francisco, California
| | - Christopher C Dvorak
- Allergy Immunology and Blood and Marrow Transplant Division at Benioff Children's Hospital at University of California San Francisco, San Francisco, California
| | - Denice Kong
- Immunogenetics and Transplantation Laboratory, University of California San Francisco, San Francisco, California
| | - Jimmy Hwang
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Jueleah Expose-Spencer
- Allergy Immunology and Blood and Marrow Transplant Division at Benioff Children's Hospital at University of California San Francisco, San Francisco, California
| | - Michael Gates
- Department of Blood and Marrow Transplantation, All Children's Hospital, St. Petersburg, Florida
| | - Morton J Cowan
- Allergy Immunology and Blood and Marrow Transplant Division at Benioff Children's Hospital at University of California San Francisco, San Francisco, California
| |
Collapse
|
86
|
Shalabi H, Angiolillo A, Fry TJ. Beyond CD19: Opportunities for Future Development of Targeted Immunotherapy in Pediatric Relapsed-Refractory Acute Leukemia. Front Pediatr 2015; 3:80. [PMID: 26484338 PMCID: PMC4589648 DOI: 10.3389/fped.2015.00080] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 09/18/2015] [Indexed: 12/30/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has been used as a targeted approach in cancer therapy. Relapsed and refractory acute leukemia in pediatrics has been difficult to treat with conventional therapy due to dose-limiting toxicities. With the recent success of CD 19 CAR in pediatric patients with B cell acute lymphoblastic leukemia (ALL), this mode of therapy has become a very attractive option for these patients with high-risk disease. In this review, we will discuss current treatment paradigms of pediatric acute leukemia and potential therapeutic targets for additional high-risk populations, including T cell ALL, AML, and infant ALL.
Collapse
Affiliation(s)
- Haneen Shalabi
- Center for Cancer and Blood Disorders, Children's National Medical Center , Washington, DC , USA
| | - Anne Angiolillo
- Center for Cancer and Blood Disorders, Children's National Medical Center , Washington, DC , USA
| | - Terry J Fry
- Hematologic Malignancies Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA
| |
Collapse
|
87
|
Ziakas PD, Zervou FN, Zacharioudakis IM, Mylonakis E. Graft-versus-host disease prophylaxis after transplantation: a network meta-analysis. PLoS One 2014; 9:e114735. [PMID: 25485632 PMCID: PMC4259365 DOI: 10.1371/journal.pone.0114735] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 11/13/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Graft-versus-host Disease (GvHD) prophylaxis after allogeneic hematopoietic stem-cell transplantation (HSCT) is an ongoing effort but relative effects of different policies are not systematically explored. METHODS We systematically reviewed 30-year evidence on GvHD prophylaxis and quantified the relative effect of different policies using a network meta-analysis. We searched PubMed and the Cochrane Library for randomized studies on the topic. The primary outcome of interest was grade II-IV acute GvHD over 0 or I (with odds ratio OR <1 denoting benefit). FINDINGS Thirty-three eligible studies that enrolled 3,440 patients (published up to June 2014), provided data on seven immunosuppressive drugs namely cyclosporin A (CsA), methotrexate (MTX), anti-thymocyte globulin (ATG), mycophenolate mofetil (MMF), tacrolimus, sirolimus or corticosteroids and their combinations to calculate 14 direct and 21 indirect effects. The majority of trials (32/33) referred to myeloablative conditioning and sibling transplants (25/33). Tacrolimus/MTX (OR 0.44; 95% 0.27-0.70, number needed to treat to benefit, i.e. to avert a case of II-IV GvHD, NNTB = 5) and ATG/CsA/MTX (OR 0.45; 95%CI 0.26-0.78; NNTB = 5) were superior over CsA/MTX. ATG/CsA/MTX did not differ from tacrolimus/MTX (indirect evidence). Sirolimus-based prophylaxis outperformed CsA/MTX (OR 0.10; 95%CI 0.02-0.49, NNTB = 4) and marginally outperformed tacrolimus/MTX (OR 0.22; 95%CI 0.05-1.11). Add-on corticosteroids had no benefit over CsA/MTX. CONCLUSIONS Tacrolimus/MTX and ATG/CsA/MTX were the outperformers over CsA/MTX, but sirolimus-based regimens showed also potential. More randomized data are needed for reduced-intensity conditioning, as well as for MMF and sirolimus-containing regimens.
Collapse
Affiliation(s)
- Panayiotis D. Ziakas
- Division of Infectious Diseases, Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Fainareti N. Zervou
- Division of Infectious Diseases, Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Ioannis M. Zacharioudakis
- Division of Infectious Diseases, Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
88
|
Pulsipher MA. Chimerism versus Minimal Residual Disease Monitoring after Allogeneic Transplantation—When Do We Act and Will Intervention Improve Outcomes? Biol Blood Marrow Transplant 2014; 20:1461-2. [DOI: 10.1016/j.bbmt.2014.07.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 07/25/2014] [Indexed: 11/26/2022]
|
89
|
Bleakley M, Kean L. Future of allogeneic hematopoietic stem cell transplantation for chemotherapy-resistant pediatric acute leukemia: potential advances. Int J Hematol Oncol 2014. [DOI: 10.2217/ijh.14.37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Marie Bleakley
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- University of Washington Department of Pediatrics, Seattle, WA, USA
| | - Leslie Kean
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- University of Washington Department of Pediatrics, Seattle, WA, USA
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
| |
Collapse
|
90
|
Relapse post hematopoietic SCT remains the Achilles heel for the field. Bone Marrow Transplant 2014; 49:997-8. [PMID: 25096916 DOI: 10.1038/bmt.2014.134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
91
|
Kornblit B, Maloney DG, Storer BE, Maris MB, Vindeløv L, Hari P, Langston AA, Pulsipher MA, Bethge WA, Chauncey TR, Lange T, Petersen FB, Hübel K, Woolfrey AE, Flowers MED, Storb R, Sandmaier BM. A randomized phase II trial of tacrolimus, mycophenolate mofetil and sirolimus after non-myeloablative unrelated donor transplantation. Haematologica 2014; 99:1624-31. [PMID: 25085357 DOI: 10.3324/haematol.2014.108340] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The study is a randomized phase II trial investigating graft-versus-host disease prophylaxis after non-myeloablative (90 mg/m(2) fludarabine and 2 Gy total body irradiation) human leukocyte antigen matched unrelated donor transplantation. Patients were randomized as follows: arm 1 - tacrolimus 180 days and mycophenolate mofetil 95 days (n=69); arm 2 - tacrolimus 150 days and mycophenolate mofetil 180 days (n=71); arm 3 - tacrolimus 150 days, mycophenolate mofetil 180 days and sirolimus 80 days (n=68). All patients had sustained engraftment. Grade II-IV acute graft-versus-host disease rates in the 3 arms were 64%, 48% and 47% at Day 150, respectively (arm 3 vs. arm 1 (hazard ratio 0.62; P=0.04). Owing to the decreased incidence of acute graft-versus-host disease, systemic steroid use was lower at Day 150 in arm 3 (32% vs. 55% in arm 1 and 49% in arm 2; overall P=0.009 by hazard ratio analysis). The Day 150 incidence of cytomegalovirus reactivation was lower in arm 3 (arm 1, 54%; arm 2, 47%; arm 3, 22%; overall P=0.002 by hazard ratio analysis). Non-relapse mortality was comparable in the three arms at two years (arm 1, 26%; arm 2, 23%; arm 3, 18%). Toxicity rates and other outcome measures were similar between the three arms. The addition of sirolimus to tacrolimus and mycophenolate mofetil is safe and associated with lower incidence of acute graft-versus-host disease and cytomegalovirus reactivation. (clinicaltrials.gov identifier: 00105001).
Collapse
Affiliation(s)
- Brian Kornblit
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - David G Maloney
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA Division of Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Barry E Storer
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA Department of Biostatistics, University of Washington, Seattle, WA, USA
| | | | | | | | | | - Michael A Pulsipher
- Division of Hematology and Hematologic Malignancies, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | | | | | | | | | - Ann E Woolfrey
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA Division of Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mary E D Flowers
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA Division of Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Rainer Storb
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA Division of Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Brenda M Sandmaier
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA Division of Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
92
|
Tacrolimus/sirolimus vs tacrolimus/methotrexate as GVHD prophylaxis after matched, related donor allogeneic HCT. Blood 2014; 124:1372-7. [PMID: 24982504 DOI: 10.1182/blood-2014-04-567164] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Grades 2-4 acute graft-versus-host disease (GVHD) occurs in approximately 35% of matched, related donor (MRD) allogeneic hematopoietic cell transplantation (HCT) recipients. We sought to determine if the combination of tacrolimus and sirolimus (Tac/Sir) was more effective than tacrolimus and methotrexate (Tac/Mtx) in preventing acute GVHD and early mortality after allogeneic MRD HCT in a phase 3, multicenter trial. The primary end point of the trial was to compare 114-day grades 2-4 acute GVHD-free survival using an intention-to-treat analysis of 304 randomized subjects. There was no difference in the probability of day 114 grades 2-4 acute GVHD-free survival (67% vs 62%, P = .38). Grades 2-4 GVHD was similar in the Tac/Sir and Tac/Mtx arms (26% vs 34%, P = .48). Neutrophil and platelet engraftment were more rapid in the Tac/Sir arm (14 vs 16 days, P < .001; 16 vs 19 days, P = .03). Oropharyngeal mucositis was less severe in the Tac/Sir arm (peak Oral Mucositis Assessment Scale score 0.70 vs 0.96, P < .001), but otherwise toxicity was similar. Chronic GVHD, relapse-free survival, and overall survival at 2 years were no different between study arms (53% vs 45%, P = .06; 53% vs 54%, P = .77; and 59% vs 63%, P = .36). Based on similar long-term outcomes, more rapid engraftment, and less oropharyngeal mucositis, the combination of Tac/Sir is an acceptable alternative to Tac/Mtx after MRD HCT. This study was funded by the National Heart, Lung, and Blood Institute and the National Cancer Institute; and the trial was registered at www.clinicaltrials.gov as #NCT00406393.
Collapse
|
93
|
Pulsipher MA, Wayne AS, Schultz KR. New frontiers in pediatric Allo-SCT: novel approaches for children and adolescents with ALL. Bone Marrow Transplant 2014; 49:1259-65. [PMID: 24933210 DOI: 10.1038/bmt.2014.114] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 02/06/2014] [Accepted: 02/13/2014] [Indexed: 11/09/2022]
Abstract
Although most children with ALL can be cured by chemotherapy approaches, allogeneic hematopoietic cell transplant (HCT) therapy offers a better chance of cure to selected high-risk patients in first remission and most children who relapse. Although transplant-related mortality has decreased significantly in the past decade, relapse remains high after HCT for ALL; developing strategies to decrease relapse and improve survival are vital. Recent studies have shown that relapse risk can be accurately defined using measurements of minimal residual disease (MRD) both pre- and post-HCT and by knowing whether patients get GVHD in the first 2 months after transplant. With these risk definitions in hand, investigators are now applying novel agents and immunotherapeutic methods in attempt to lower MRD before transplant and modulate the GVL effect after transplant. With powerful new immunological approaches coming on line, the transplant process itself will likely expand to include pre and/or post-HCT interventions aimed at reducing relapse.
Collapse
Affiliation(s)
- M A Pulsipher
- Division of Hematology and Hematological Malignancies, Primary Children's Hospital, University of Utah School of Medicine/Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - A S Wayne
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, The Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - K R Schultz
- Department of Pediatrics, University of BC, BC Children's Hospital, Vancouver, British Columbia, Canada
| |
Collapse
|
94
|
Talano JM, Pulsipher MA, Symons HJ, Militano O, Shereck EB, Giller RH, Hancock L, Morris E, Cairo MS. New frontiers in pediatric Allo-SCT. Bone Marrow Transplant 2014; 49:1139-45. [PMID: 24820213 DOI: 10.1038/bmt.2014.89] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 03/13/2014] [Accepted: 03/15/2014] [Indexed: 01/02/2023]
Abstract
The inaugural meeting of 'New Frontiers in Pediatric Allogeneic Stem Cell Transplantation' organized by the Pediatric Blood and Transplant Consortium (PBMTC) was held at the American Society of Pediatric Hematology and Oncology Annual Meeting. This meeting provided an international platform for physicians and investigators active in the research and utilization of pediatric Allo-SCT in children and adolescents with malignant and non-malignant disease (NMD), to share information and develop future collaborative strategies. The primary objectives of the conference included: (1) to present advances in Allo-SCT in pediatric ALL and novel pre and post-transplant immunotherapy; (2) to highlight new strategies in alternative allogeneic stem cell donor sources for children and adolescents with non-malignant hematological disorders; (3) to discuss timing of immune reconstitution after Allo-SCT and methods of facilitating more rapid recovery of immunity; (4) to identify strategies of utilizing Allo-SCT in pediatric myeloproliferative disorders; (5) to develop diagnostic and therapeutic approaches to hematological complications post pediatric Allo-SCT; (6) to enhance the understanding of new novel cellular therapeutic approaches to pediatric malignant and non-malignant hematological disorders; and (7) to discuss optimizing drug therapy in pediatric recipients of Allo-SCT. This paper will provide a brief overview of the conference.
Collapse
Affiliation(s)
- J M Talano
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - M A Pulsipher
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - H J Symons
- Department of Oncology, Johns Hopkins Hospital, Baltimore, MD, USA
| | - O Militano
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | - E B Shereck
- Oregon Health and Science University, Portland, OR, USA
| | - R H Giller
- Children's Hospital Colorado, Aurora, CO, USA
| | - L Hancock
- Pediatric Blood and Marrow Transplant Consortium, Monrovia, CA, USA
| | - E Morris
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | - M S Cairo
- 1] Department of Pediatrics, New York Medical College, Valhalla, NY, USA [2] Department of Medicine, New York Medical College, Valhalla, NY, USA [3] Department of Pathology, New York Medical College, Valhalla, NY, USA [4] Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA [5] Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| |
Collapse
|