1
|
Deng L, Yu X, Song X, Guan R, Li W, Liu X, Shao Y, Hou Y, Zhao Y, Wang J, Liu Y, Xiao Q, Xin B, Zhou F. The prophylactic application of low-dose rabbit antithymocyte globulin in matched siblings HSCT with high-risk factors for graft-versus-host disease. Transpl Immunol 2024; 87:102131. [PMID: 39307438 DOI: 10.1016/j.trim.2024.102131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/13/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
Relapse and graft-versus-host disease (GVHD) are currently the predominant causes of mortality post allogeneic hematopoietic stem cell transplantation (allo-HSCT). The contentious use of antithymocyte globulin (ATG) for preventing GVHD in matched sibling HSCT scenarios has been a topic of significant debate. A retrospective analysis was conducted on matched sibling HSCT cases with high-risk factors for GVHD in our center from January 2018 to June 2023. Our assessment revealed that the group administered with ATG exhibited a 30 % incidence of acute GVHD (aGVHD), in contrast to 81.8 % in the non-ATG cohort (P = 0.037) among matched sibling HSCT cases with high GVHD risk factors. Furthermore, chronic GVHD (cGVHD) occurred in 20 % of the ATG group and 72.7 % of the non-ATG group (P = 0.03). Notably, the administration of ATG did not significantly impact disease relapse (p = 0.149), infection rates (p = 0.64), granulocyte recovery time (p = 0.15), platelet recovery time (p = 0.12), overall survival (p = 0.889), or disease-free survival time (p = 0.787). The use of rabbit antithymocyte globulin (r-ATG) at a 5 mg/kg dosage demonstrated a notable reduction in aGVHD and cGVHD incidences within sibling matched HSCT cases with high-risk factors for GVHD, without increasing rates of disease recurrence or infections. These findings highlight the potential benefit of using low-dose r-ATG in high-risk of GVHD sibling matched allogeneic HSCTs, although further validation with a larger cohort is necessary.
Collapse
Affiliation(s)
- Lei Deng
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Xiaolin Yu
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Xiaocheng Song
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Rui Guan
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Wenjun Li
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Ximing Liu
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Yan Shao
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Yixi Hou
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Yuerong Zhao
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Jing Wang
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Yue Liu
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Qianqian Xiao
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Bo Xin
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Fang Zhou
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China.
| |
Collapse
|
2
|
Muffly L, Liang EC, Dolan JG, Pulsipher MA. How I use next-generation sequencing-MRD to plan approach and prevent relapse after HCT for children and adults with ALL. Blood 2024; 144:253-261. [PMID: 38728375 PMCID: PMC11302453 DOI: 10.1182/blood.2023023699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/11/2024] [Accepted: 04/24/2024] [Indexed: 05/12/2024] Open
Abstract
ABSTRACT Measurable residual disease (MRD) evaluation by multiparameter flow cytometry (MFC) or quantitative polymerase chain reaction methods is an established standard of care for assessing risk of relapse before or after hematopoietic cell transplantation (HCT) for acute lymphoblastic leukemia (ALL). Next-generation sequencing (NGS)-MRD has emerged as a highly effective approach that allows for the detection of lymphoblasts at a level of <1 in 106 nucleated cells, increasing sensitivity of ALL detection by 2 to 3 logs. Early studies have shown superior results compared with MFC and suggest that NGS-MRD may allow for the determination of patients in whom reduced toxicity transplant preparative approaches could be deployed without sacrificing outcomes. Many centers/study groups have implemented immune modulation approaches based on MRD measurements that have resulted in improved outcomes. Challenges remain with NGS-MRD, because it is not commercially available in many countries, and interpretation of results can be complex. Through patient case review, discussion of relevant studies, and detailed expert opinion, we share our approach to NGS-MRD testing before and after HCT in pediatric and adult ALL. Improved pre-HCT risk classification and post-HCT monitoring for relapse in bone marrow and less invasive peripheral blood monitoring by NGS-MRD may lead to alternative approaches to prevent relapse in patients undergoing this challenging procedure.
Collapse
Affiliation(s)
- Lori Muffly
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Emily C. Liang
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - J. Gregory Dolan
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Intermountain Primary Children’s Hospital, Spencer Fox Eccles School of Medicine at the University of Utah, Salt Lake City, UT
| | - Michael A. Pulsipher
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Intermountain Primary Children’s Hospital, Spencer Fox Eccles School of Medicine at the University of Utah, Salt Lake City, UT
- Division of Pediatric Hematology and Oncology, Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine at the University of Utah, Salt Lake City, UT
| |
Collapse
|
3
|
Kernan NA, Klein E, Mauguen A, Torok-Castanza J, Prockop SE, Scaradavou A, Curran K, Spitzer B, Cancio M, Ruggiero J, Allen J, Harris A, Oved J, O'Reilly RJ, Boelens JJ. Persistent or New Cytopenias Predict Relapse Better than Routine Bone Marrow Aspirate Evaluations After Hematopoietic Cell Transplantation for Acute Leukemia or Myelodysplastic Syndrome in Children and Young Adult Patients. Transplant Cell Ther 2024; 30:692.e1-692.e12. [PMID: 38643958 DOI: 10.1016/j.jtct.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/02/2024] [Accepted: 04/17/2024] [Indexed: 04/23/2024]
Abstract
The clinical value of serial routine bone marrow aspirates (rBMAs) in the first year after allogeneic hematopoietic cell transplantation (alloHCT) to detect or predict relapse of acute leukemia (AL) and myelodysplastic syndrome (MDS) in pediatric and young adult patients is unclear. The purpose of this analysis was to determine if assessment of minimal residual disease (MRD) by multiparameter flow cytometry (MFC, MFC-MRD) or donor chimerism (DC) in rBMAs or serial complete blood counts (CBCs) done in the year after alloHCT predicted relapse of AL or MDS in pediatric and young adult patients. We completed a retrospective analysis of patients with AL or MDS who had rBMAs performed after alloHCT between January 2012 and June 2018. Bone marrow (BM) was evaluated at approximately 3, 6, and 12 months for disease recurrence by morphology, MFC-MRD, and percent DC by short tandem repeat molecular testing. CBCs were performed at every clinic visit. The main outcome of interest was an assessment of whether MFC-MRD or DC in rBMAs or serial CBCs done in the year after alloHCT predicted relapse in AL or MDS pediatric and young adult patients. A total of 121 recipients with a median age of 13 years (range 1 to 32) were included: 108 with AL and, 13 with MDS. A total of 423 rBMAs (median 3; 0 to 13) were performed. Relapse at 2 years was 23% (95% CI: 16% to 31%) and at 5 years 25% (95% CI: 18% to 33%). One hundred fifty-four of 157 (98%) rBMAs evaluated for MRD by MFC were negative and did not preclude subsequent relapse. Additionally, low DC (<95%) did not predict relapse and high DC (≥95%) did not preclude relapse. For patients alive without relapse at 1 year, BM DC (P = .74) and peripheral T-cell DC (P = .93) did not predict relapse. Six patients with low-level T-cell and/or BM DC had a total of 8 to 20 BM evaluations, none of these patients relapsed. However, CBC results were informative for relapse; 28 of 31 (90%) relapse patients presented with an abnormal CBC with peripheral blood (PB) blasts (16 patients), cytopenias (9 patients), or extramedullary disease (EMD, 3 patients). Two patients with BM blasts >5% on rBMA had circulating blasts within 5 weeks of rBMA. Neutropenia (ANC <1.5 K/mcl) at 1 year was predictive of relapse (P = .01). Neutropenia and thrombocytopenia (<160 K/mcl) were predictive of disease-free survival (DFS) with inferior DFS for ANC <1.5 K/mcl, P = .001, or platelet count <160 K/mcl (P = .04). These results demonstrate rBMAs after alloHCT assessed for MRD by MFC and/or for level of DC are poor predictors for relapse in pediatric and young adult patients with AL or MDS. Relapse in these patients presents with PB blasts, cytopenias, or EMD. ANC and platelet count at 1-year were highly predictive for DFS.
Collapse
Affiliation(s)
- Nancy A Kernan
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service. New York, New York.
| | - Elizabeth Klein
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service. New York, New York
| | - Audrey Mauguen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Susan E Prockop
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service. New York, New York; Dana Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA Harvard Medical School, Boston, Massachusetts
| | - Andromachi Scaradavou
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service. New York, New York; Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Kevin Curran
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service. New York, New York; Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Barbara Spitzer
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service. New York, New York; Hackensack University Medical Center, Hackensack, New Jersey
| | - Maria Cancio
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service. New York, New York; Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Julianne Ruggiero
- Division of Nursing, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jennifer Allen
- Division of Nursing, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrew Harris
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service. New York, New York; Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Joseph Oved
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service. New York, New York; Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Richard J O'Reilly
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service. New York, New York; Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Jaap Jan Boelens
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service. New York, New York; Department of Pediatrics, Weill Cornell Medicine, New York, New York
| |
Collapse
|
4
|
Gharial J, Guilcher G, Truong T, Shah R, Desai S, Rojas-Vasquez M, Kangarloo B, Lewis V. Busulfan with 400 centigray of total body irradiation and higher dose fludarabine: An alternative regimen for hematopoietic stem cell transplantation in pediatric acute lymphoblastic leukemia. Pediatr Blood Cancer 2024; 71:e30844. [PMID: 38217082 DOI: 10.1002/pbc.30844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 12/01/2023] [Accepted: 12/21/2023] [Indexed: 01/14/2024]
Abstract
BACKGROUND Hematopoietic stem cell transplantation can be curative for children with difficult-to-treat leukemia. The conditioning regimen utilized is known to influence outcomes. We report outcomes of the conditioning regimen used at the Alberta Children's Hospital, consisting of busulfan (with pharmacokinetic target of 3750 μmol*min/L/day ±10%) for 4 days, higher dose (250 mg/m2 ) fludarabine and 400 centigray (cGy) of total body irradiation. PROCEDURE This retrospective study involved children receiving transplant for acute lymphoblastic leukemia (ALL). It compared children who fell within the target range for busulfan with those who were either not measured or were measured and fell outside this range. All other treatment factors were identical. RESULTS Twenty-nine children (17 within target) were evaluated. All subjects engrafted neutrophils with a median [interquartile range] time of 14 days [8-30 days]. The cumulative incidence of acute graft-versus-host disease was 44.8% [95% confidence interval, CI: 35.6%-54.0%], while chronic graft-versus-host disease was noted in 16.0% [95% CI: 8.7%-23.3%]. At 2 years, the overall survival was 78.1% [95% CI: 70.8%-86.4%] and event-free survival was 74.7% [95% CI: 66.4%-83.0%]. Cumulative incidence of relapse was 11.3% [95% CI: 5.1%-17.5%]. There were no statistically significant differences in between the group that received targeted busulfan compared with the untargeted group. CONCLUSION Our conditioning regiment for children with ALL resulted in outcomes comparable to standard treatment with acceptable toxicities and significant reduction in radiation dose. Targeting busulfan dose in this cohort did not result in improved outcomes.
Collapse
Affiliation(s)
- Jaspreet Gharial
- Section of Pediatric Hematology Oncology and Bone Marrow Transplantation, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Gregory Guilcher
- Section of Pediatric Hematology Oncology and Bone Marrow Transplantation, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Tony Truong
- Section of Pediatric Hematology Oncology and Bone Marrow Transplantation, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Ravi Shah
- Section of Pediatric Hematology Oncology and Bone Marrow Transplantation, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Sunil Desai
- Division of Pediatric Hematology/Oncology & Palliative Care, Stollery Children's Hospital, Edmonton, Alberta, Canada
| | - Marta Rojas-Vasquez
- Division of Pediatric Hematology/Oncology & Palliative Care, Stollery Children's Hospital, Edmonton, Alberta, Canada
| | - Bill Kangarloo
- Pharmacokinetic Scientist, Alberta Blood and Marrow Transplant Program, Foothills Hospital, and Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | - Victor Lewis
- Section of Pediatric Hematology Oncology and Bone Marrow Transplantation, Alberta Children's Hospital, Calgary, Alberta, Canada
| |
Collapse
|
5
|
Jaime-Pérez JC, Meléndez-Flores JD, Valdespino-Valdes J, Gómez-De León A, Colunga-Pedraza PR, Gutiérrez-Aguirre CH, Cantú-Rodríguez OG, Gómez-Almaguer D. Graft-versus-host disease after an outpatient peripheral blood hematopoietic cell transplant using reduced-intensity conditioning: a single-center LATAM experience. Expert Rev Hematol 2024; 17:77-86. [PMID: 38226642 DOI: 10.1080/17474086.2024.2305372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 12/26/2023] [Indexed: 01/17/2024]
Abstract
BACKGROUND HLA compatibility predicts allogeneic hematopoietic cell transplant (allo-HCT) and graft-versus-host disease (GvHD) outcomes. There is insufficient information regarding GvHD outcomes for outpatient HLA-identical and haploidentical-HCT employing reduced-intensity conditioning (RIC). RESEARCH DESIGN AND METHODS We compare GvHD outcomes between donor types and report risk factors associated with GvHD. Stem cell source was T-cell replete peripheral blood. GvHD prophylaxis was post-transplant cyclophosphamide (PT-CY), mycophenolic acid, and calcineurin inhibitors for haploidentical (n = 107) and oral cyclosporine (CsA) plus methotrexate i.v. for HLA-identical (n = 89) recipients. RESULTS One hundred and ninety-six HCT transplant patients were included. aGvHD and cGvHD frequency were similar between HCT types. aGvHD severity was comparable, but severe cGvHD was less frequent in the haploidentical group (p = .011). One-hundred-day cumulative incidence (CI) of aGvHD for haploidentical and HLA-identical was 31% and 33% (p = .84); 2-year CI of cGvHD was 32% and 38% (p = .6), respectively. Haploidentical recipients had less steroid-refractory cGvHD (p = .043). Patients with cGvHD had less 2-year relapse (p = .003); both aGvHD and cGvHD conferred higher OS (p = .010 and p = .001), respectively. Male sex was protective for steroid-refractory cGvHD (p = .028). CONCLUSIONS Acute and chronic GvHD rates were comparable between HLA-identical and haploidentical transplant groups. cGvHD severity was lower in the haploidentical group.
Collapse
Affiliation(s)
- José Carlos Jaime-Pérez
- Hematology Department, Internal Medicine Division, Dr. José Eleuterio González University Hospital, School of Medicine, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Jesús Daniel Meléndez-Flores
- Hematology Department, Internal Medicine Division, Dr. José Eleuterio González University Hospital, School of Medicine, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Jorge Valdespino-Valdes
- Hematology Department, Internal Medicine Division, Dr. José Eleuterio González University Hospital, School of Medicine, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Andrés Gómez-De León
- Hematology Department, Internal Medicine Division, Dr. José Eleuterio González University Hospital, School of Medicine, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Perla Rocío Colunga-Pedraza
- Hematology Department, Internal Medicine Division, Dr. José Eleuterio González University Hospital, School of Medicine, Universidad Autónoma de Nuevo León, Monterrey, México
| | - César Homero Gutiérrez-Aguirre
- Hematology Department, Internal Medicine Division, Dr. José Eleuterio González University Hospital, School of Medicine, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Olga Graciela Cantú-Rodríguez
- Hematology Department, Internal Medicine Division, Dr. José Eleuterio González University Hospital, School of Medicine, Universidad Autónoma de Nuevo León, Monterrey, México
| | - David Gómez-Almaguer
- Hematology Department, Internal Medicine Division, Dr. José Eleuterio González University Hospital, School of Medicine, Universidad Autónoma de Nuevo León, Monterrey, México
| |
Collapse
|
6
|
Shyr D, Davis KL, Bertaina A. Stem cell transplantation for ALL: you've always got a donor, why not always use it? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:84-90. [PMID: 38066901 PMCID: PMC10726989 DOI: 10.1182/hematology.2023000423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Hematopoietic stem cell transplantation (HSCT) represents a consolidated therapeutic strategy for high-risk pediatric acute lymphoblastic leukemia (ALL), offering the potential for curative treatment. This manuscript delves into the debate around the more universal application of HSCT for pediatric ALL in the modern era, considering the ubiquitous availability of suitable donors. In fact, despite significant advancements in chemotherapy, targeted therapy, and immunotherapy, a subset of pediatric patients with ALL with high-risk features or relapse continue to encounter poor prognostic outcomes. For this subgroup of patients, HSCT often remains the only potentially curative measure, leveraging the graft-versus- leukemia effect for long-term disease control. Nevertheless, the procedure's complexity and associated risks have traditionally curtailed its widespread use. However, the scenario is shifting with improvements in HLA matching, availability of alternative donor sources, less toxic conditioning regimens, and improved supportive care protocols. Concurrently, emerging therapies like CD19+ CAR T cells present new considerations for definitive therapy selection in relapsed/ refractory ALL. This article reviews critical current evidence and debates the potential of HSCT as a more universal treatment for ALL, reevaluating traditional treatment stratification in light of the constant availability of stem cell donors.
Collapse
Affiliation(s)
- David Shyr
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA
- Center for Definitive and Curative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA
| | - Kara L Davis
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA
- Center for Cancer Cellular Therapy, Stanford University School of Medicine, Palo Alto, CA
| | - Alice Bertaina
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA
- Center for Definitive and Curative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA
| |
Collapse
|
7
|
Prockop S, Wachter F. The current landscape: Allogeneic hematopoietic stem cell transplant for acute lymphoblastic leukemia. Best Pract Res Clin Haematol 2023; 36:101485. [PMID: 37611999 DOI: 10.1016/j.beha.2023.101485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 05/31/2023] [Indexed: 08/25/2023]
Abstract
One of the consistent features in development of hematopoietic stem cell transplant (HCT) for Acute Lymphoblastic Leukemia (ALL) is the rapidity with which discoveries in the laboratory are translated into innovations in clinical care. Just a few years after murine studies demonstrated that rescue from radiation induced marrow failure is mediated by cellular not humoral factors, E. Donnall Thomas reported on the transfer of bone marrow cells into irradiated leukemia patients. This was followed quickly by the first descriptions of Graft versus Leukemia (GvL) effect and Graft versus Host Disease (GvHD). Despite the pivotal nature of these findings, early human transplants were uniformly unsuccessful and identified the challenges that continue to thwart transplanters today - leukemic relapse, regimen related toxicity, and GvHD. While originally only an option for young, fit patients with a matched family donor, expansion of the donor pool to include unrelated donors, umbilical cord blood units, and more recently the growing use of haploidentical donors have all made transplant a more accessible therapy for patients with ALL. Novel agents for conditioning, prevention and treatment of GvHD have improved outcomes and investigators continue to develop novel treatment strategies that balance regimen related toxicity with disease control. Our evolving understanding of how to prevent and treat GvHD and how to prevent relapse are incorporated into novel clinical trials that are expected to further improve outcomes. Here we review current considerations and future directions for both adult and pediatric patients undergoing HCT for ALL, including indication for transplant, donor selection, cytoreductive regimens, and outcomes.
Collapse
Affiliation(s)
- Susan Prockop
- Pediatric Stem Cell Transplant Program, DFCI/BCH Center for Cancer and Blood Disorders, Pediatrics, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States.
| | - Franziska Wachter
- Pediatric Stem Cell Transplant Program, DFCI/BCH Center for Cancer and Blood Disorders, Pediatrics, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States.
| |
Collapse
|
8
|
Kitko CL, Bollard CM, Cairo MS, Chewning J, Fry TJ, Pulsipher MA, Shenoy S, Wall DA, Levine JE. Children's Oncology Group's 2023 blueprint for research: Cellular therapy and stem cell transplantation. Pediatr Blood Cancer 2023; 70 Suppl 6:e30577. [PMID: 37480158 PMCID: PMC10527977 DOI: 10.1002/pbc.30577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/23/2023]
Abstract
Since the publication of the last Cellular Therapy and Stem Cell Transplant blueprint in 2013, Children's Oncology Group cellular therapy-based trials advanced the field and created new standards of care across a wide spectrum of pediatric cancer diagnoses. Key findings include that tandem autologous transplant improved survival for patients with neuroblastoma and atypical teratoid/rhabdoid brain tumors, one umbilical cord blood (UCB) donor was safer than two UCB donors, killer immunoglobulin receptor (KIR) mismatched donors did not improve survival for pediatric acute myeloid leukemia when in vivo T-cell depletion is used, and the depth of remission as measured by next-generation sequencing-based minimal residual disease assessment pretransplant was the best predictor of relapse for acute lymphoblastic leukemia. Plans for the next decade include optimizing donor selection for transplants for acute leukemia/myelodysplastic syndrome, using novel engineered cellular therapies to target a wide array of malignancies, and developing better treatments for cellular therapy toxicities such as viral infections and graft-vs-host disease.
Collapse
Affiliation(s)
- Carrie L. Kitko
- Pediatric Stem Cell Transplant Program, Vanderbilt University Medical Center, Nashville, TN
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC
- GW Cancer Center, George Washington University, Washington, DC
- Division of Blood and Marrow Transplantation, Children’s National Hospital, Washington, DC
| | - Mitchell S. Cairo
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Maria Fareri Children's Hospital, Westchester Medical Center, New York Medical College, Valhalla, New York, NY
| | - Joseph Chewning
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL
| | - Terry J. Fry
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO
| | - Michael A. Pulsipher
- Division of Hematology and Oncology, Intermountain Primary Children’s Hospital, Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine, Salt Lake City, UT
| | - Shalini Shenoy
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Washington University, St Louis, MO
| | - Donna A. Wall
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Canada
| | - John E. Levine
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
9
|
Talleur AC, Pui CH, Karol SE. What is Next in Pediatric B-cell Precursor Acute Lymphoblastic Leukemia. LYMPHATICS 2023; 1:34-44. [PMID: 38269058 PMCID: PMC10804398 DOI: 10.3390/lymphatics1010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Cure rates now exceed 90% in many contemporary trials for children with B-cell acute lymphoblastic leukemia (ALL). However, treatment remains suboptimal and therapy is toxic for all patients. New treatment options potentially offer the chance to reduce both treatment resistance and toxicity. Here, we review recent advances in ALL diagnostics, chemotherapy, and immunotherapy. In addition to describing recently published results, we also attempt to project the impact of these new developments into the future to imagine what B-ALL therapy may look like in the next few years.
Collapse
Affiliation(s)
- Aimee C Talleur
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Seth E Karol
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
10
|
Varadarajan I, Pierce E, Scheuing L, Morris A, El Chaer F, Keng M. Post-Hematopoietic Cell Transplantation Relapsed Acute Lymphoblastic Leukemia: Current Challenges and Future Directions. Onco Targets Ther 2023; 16:1-16. [PMID: 36685611 PMCID: PMC9849790 DOI: 10.2147/ott.s274551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 12/14/2022] [Indexed: 01/15/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) represents an important and potentially curative treatment option for adult patients with acute lymphoblastic leukemia. Relapse continues to remain the most important factor influencing overall survival post allo-HCT. We discuss early identification, clinical manifestations, and management of relapsed disease. Routine evaluation of measurable residual disease (MRD) and change in donor chimerism play a crucial role in early detection. Pivotal clinical trials have led to FDA approval of multiple novel agents like blinatumomab and inotuzumab. Combining targeted therapy with cellular immunotherapy serves as the backbone for prolonging overall survival in these patients. Donor lymphocyte infusions have traditionally been used in relapsed disease with suboptimal outcomes. This review provides insight into use of cellular therapy in MRD positivity and decreasing donor chimerism. It also discusses various modalities of combining cellular therapy with novel agents and discussing the impact of chimeric antigen receptor T-cell therapy in the setting of post allo-HCT relapse both as consolidative therapy and as a bridge to second transplant.
Collapse
Affiliation(s)
- Indumathy Varadarajan
- Department of Medicine, Division of Hematology and Oncology, University of Virginia, Charlottesville, VA, USA
| | - Eric Pierce
- Department of Medicine, Division of Hematology and Oncology, University of Virginia, Charlottesville, VA, USA
| | - Lisa Scheuing
- Department of Medicine, Division of Hematology and Oncology, University of Virginia, Charlottesville, VA, USA
| | - Amy Morris
- Department of Pharmacy Services, University of Virginia, Charlottesville, VA, USA
| | - Firas El Chaer
- Department of Medicine, Division of Hematology and Oncology, University of Virginia, Charlottesville, VA, USA
| | - Michael Keng
- Department of Medicine, Division of Hematology and Oncology, University of Virginia, Charlottesville, VA, USA,Correspondence: Michael Keng, Division of Hematology & Oncology, University of Virginia Comprehensive Cancer Center, West Complex Room 6009, 1300 Jefferson Park Ave, PO Box 800716, Charlottesville, VA, 22908, USA, Tel +1 434 924 4257, Fax +1 434- 243 6068, Email
| |
Collapse
|
11
|
Moreno C, Ramos-Elbal E, Velasco P, Aguilar Y, Gonzáález Martínez B, Fuentes C, Molinos Á, Guerra-García P, Palomo P, Verdu J, Adán Pedroso RM, Vagace JM, López-Duarte M, Regueiro A, Tasso M, Dapena JL, Salinas JA, Navarro S, Bautista F, Lassaletta Á, Lendínez F, Rives S, Pascual A, Rodríguez A, Pérez-Hurtado JM, Fernández JM, Pérez-Martínez A, González-Vicent M, Díaz de Heredia C, Fuster JL. Haploidentical vs. HLA-matched donor hematopoietic stem-cell transplantation for pediatric patients with acute lymphoblastic leukemia in second remission: A collaborative retrospective study of the Spanish Group for Bone Marrow Transplantation in Children (GETMON/GETH) and the Spanish Childhood Relapsed ALL Board (ReALLNet). Front Pediatr 2023; 11:1140637. [PMID: 37020654 PMCID: PMC10067875 DOI: 10.3389/fped.2023.1140637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/23/2023] [Indexed: 04/07/2023] Open
Abstract
Introduction Studies addressing the role of haploidentical as alternative to HLA-matched donors for stem cell transplantation (SCT) often include patients with diverse hematological malignancies in different remission statuses. Methods We compared outcomes of children with acute lymphoblastic leukemia (ALL) undergoing SCT in second complete remission (CR2) from haploidentical (n = 25) versus HLA-matched donor (n = 51). Results Patients were equally distributed across both groups according to age, immunophenotype, time to and site of relapse, relapse risk-group allocation, and minimal residual disease (MRD) before SCT. Incidence of graft failure, acute graft versus host disease (GVHD), and other early complications did not differ between both groups. We found no differences in overall survival (58.7% versus 59.5%; p = .8), leukemia free survival (LFS) (48% versus 36.4%; p = .5), event free survival (40% versus 34.4%; p = .69), cumulative incidence (CI) of subsequent relapse (28% versus 40.9%; p = .69), treatment related mortality (24% versus 23.6%; p = .83), CI of cGVHD (4.5% versus 18.7%; p = .2), and chronic GVHD-free and leukemia-free survival (44% versus 26.3%; p = .3) after haploidentical donor SCT. Chronic GVHD (HR = 0.09; p=.02) had protective impact, and MRD ≥ 0.01% before SCT (HR = 2.59; p=.01) had unfavorable impact on LFS. Discussion These results support the role of haploidentical donor SCT in children with ALL in CR2.
Collapse
Affiliation(s)
- Celia Moreno
- Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | | | - Pablo Velasco
- Hospital Universitario Vall d’Hebron, Vall d’Hebron Institut de Recerca, Barcelona, Spain
| | | | - Berta Gonzáález Martínez
- Hospital Universitario La Paz, IdiPAZ, Instituto de Investigación, Hospital Universitario La Paz, Madrid, Spain
| | | | | | - Pilar Guerra-García
- Hospital Universitario La Paz, IdiPAZ, Instituto de Investigación, Hospital Universitario La Paz, Madrid, Spain
- Hospital Universitario 12 de octubre, Madrid, Spain
| | - Pilar Palomo
- Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Jaime Verdu
- Hospital Universitario de Valencia, Valencia, Spain
| | | | | | - Mónica López-Duarte
- Hospital de Valdecilla, Instituto de Investigación Sanitaria Valdecilla, IDIVAL, Santander, Spain
| | - Alexandra Regueiro
- Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
| | - María Tasso
- Hospital General Universitario Doctor Balmis, Alicante, Spain
| | - José Luis Dapena
- Pediatric Cancer Center, Hospital Sant Joan de Déu, Barcelona, Spain
- Institut de Recerca San Joan de Déu,Barcelona, Spain
| | | | - Samuel Navarro
- Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | | | | | | | - Susana Rives
- Pediatric Cancer Center, Hospital Sant Joan de Déu, Barcelona, Spain
- Institut de Recerca San Joan de Déu,Barcelona, Spain
| | | | | | | | | | - Antonio Pérez-Martínez
- Hospital Universitario La Paz, IdiPAZ, Instituto de Investigación, Hospital Universitario La Paz, Madrid, Spain
| | | | | | - José Luis Fuster
- Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
- Correspondence: José Luis Fuster
| |
Collapse
|
12
|
Long non-coding RNA signatures and related signaling pathway in T-cell acute lymphoblastic leukemia. Clin Transl Oncol 2022; 24:2081-2089. [DOI: 10.1007/s12094-022-02886-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 07/04/2022] [Indexed: 11/25/2022]
|
13
|
Hussain Y, Abdullah, Alsharif KF, Aschner M, Theyab A, Khan F, Saso L, Khan H. Therapeutic Role of Carotenoids in Blood Cancer: Mechanistic Insights and Therapeutic Potential. Nutrients 2022; 14:1949. [PMID: 35565917 PMCID: PMC9104383 DOI: 10.3390/nu14091949] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/25/2022] [Accepted: 04/29/2022] [Indexed: 02/07/2023] Open
Abstract
Blood cancers are characterized by pathological disorders causing uncontrolled hematological cell division. Various strategies were previously explored for the treatment of blood cancers, including chemotherapy, Car-T therapy, targeting chimeric antigen receptors, and platelets therapy. However, all these therapies pose serious challenges that limit their use in blood cancer therapy, such as poor metabolism. Furthermore, the solubility and stability of anticancer drugs limit efficacy and bio-distribution and cause toxicity. The isolation and purification of natural killer cells during Car-T cell therapy is a major challenge. To cope with these challenges, treatment strategies from phyto-medicine scaffolds have been evaluated for blood cancer treatments. Carotenoids represent a versatile class of phytochemical that offer therapeutic efficacy in the treatment of cancer, and specifically blood cancer. Carotenoids, through various signaling pathways and mechanisms, such as the activation of AMPK, expression of autophagy biochemical markers (p62/LC3-II), activation of Keap1-Nrf2/EpRE/ARE signaaling pathway, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), increased level of reactive oxygen species, cleaved poly (ADP-ribose) polymerase (c-PARP), c-caspase-3, -7, decreased level of Bcl-xL, cycle arrest at the G0/G1 phase, and decreasing STAT3 expression results in apoptosis induction and inhibition of cancer cell proliferation. This review article focuses the therapeutic potential of carotenoids in blood cancers, addressing various mechanisms and signaling pathways that mediate their therapeutic efficacy.
Collapse
Affiliation(s)
- Yaseen Hussain
- Lab of Controlled Release and Drug Delivery System, College of Pharmaceutical Sciences, Soochow University, Suzhou 215000, China;
- Department of Pharmacy, Bashir Institute of Health Sciences, Bharakahu, Islamabad 44000, Pakistan
| | - Abdullah
- Department of Pharmacy, University of Malakand, Chakdara 18800, Pakistan;
| | - Khalaf F. Alsharif
- Department of Clinical Laboratory, College of Applied Medical Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10463, USA;
| | - Abdulrahman Theyab
- Department of Laboratory and Blood Bank, Security Forces Hospital, P.O. Box 14799, Mecca 21955, Saudi Arabia;
- College of Medicine, Al-Faisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
| | - Fazlullah Khan
- Faculty of Pharmacy, Capital University of Science & Technology, Islamabad 44000, Pakistan;
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University, 00185 Rome, Italy;
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| |
Collapse
|
14
|
Andolina JR, Wang YC, Ji L, Freyer DR, Levine JE, Pulsipher MA, Gamis AS, Aplenc R, Roth ME, Harrison L, Cairo MS. Adolescent and young adult (AYA) versus pediatric patients with acute leukemia have a significantly increased risk of acute GVHD following unrelated donor (URD) stem cell transplantation (SCT): the Children's Oncology Group experience. Bone Marrow Transplant 2022; 57:445-452. [PMID: 34992254 PMCID: PMC9621326 DOI: 10.1038/s41409-021-01558-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/28/2021] [Accepted: 12/17/2021] [Indexed: 11/09/2022]
Abstract
Adolescent and young adult (AYA) patients with acute leukemia (AL) have inferior outcomes in comparison to younger patients, and are more likely to develop acute and chronic GVHD than younger children following HLA matched sibling donor stem cell transplant (SCT). We compared the incidence of grade II-IV acute GVHD, chronic GVHD, and survival in AYA (age 13-21 years) to younger children (age 2-12 years) who received an unrelated donor SCT for acute leukemia on Children's Oncology Group trials between 2004-2017. One hundred and eighty-eight children and young adults ages 2-21 years underwent URD SCT. Sixty-three percent were aged 2-12 and 37% were age 13-21. Older age was a risk factor for grade II-IV acute GVHD in multivariate analysis with a hazard ratio (HR) of 1.95 [95% confidence interval (CI) 1.23-3.10], but not for chronic GVHD, HR 1.25 [95% CI 0.57-2.71]. Younger patients relapsed more often (34.5 ± 4.4% vs. 22.8 ± 4.0%, p = 0.032), but their Event-Free Survival (42.6 ± 4.7% vs. 51.8 ± 6.1%, p = 0.18) and Overall Survival at 5 years (48.5 ± 4.9% vs. 51.5 ± 6.4%, p = 0.56) were not different than AYA patients. AYA patients who receive an URD SCT for acute leukemia are significantly more likely to develop grade II-IV acute GVHD, though survival is similar.
Collapse
Affiliation(s)
- Jeffrey R Andolina
- Department of Pediatrics, Golisano Children's Hospital, University of Rochester Medical Center, Rochester, NY, USA.
| | | | - Lingyun Ji
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - David R Freyer
- Transplantation and Cellular Therapy Section, Children's Hospital Los Angeles Cancer and Blood Disease Institute, USC Keck School of Medicine, Los Angeles, CA, USA
| | - John E Levine
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael A Pulsipher
- Transplantation and Cellular Therapy Section, Children's Hospital Los Angeles Cancer and Blood Disease Institute, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Alan S Gamis
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, USA
| | - Richard Aplenc
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Michael E Roth
- Department of Pediatrics, MD Anderson Cancer Center, Houston, TX, USA
| | - Lauren Harrison
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA.
- Departments of Pediatrics, Medicine, Pathology, Microbiology and Immunology, Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA.
| |
Collapse
|
15
|
Kállay KM, Algeri M, Buechner J, Krauss AC. Bispecific Antibodies and Other Non-CAR Targeted Therapies and HSCT: Decreased Toxicity for Better Transplant Outcome in Paediatric ALL? Front Pediatr 2022; 9:795833. [PMID: 35252074 PMCID: PMC8889254 DOI: 10.3389/fped.2021.795833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/15/2021] [Indexed: 11/16/2022] Open
Abstract
This review will address the place of innovative, non-chemotherapy, non-CAR-T targeted therapies in the treatment of Acute Lymphoblastic Leukaemia (ALL), focusing on their use in the hematopoietic stem cell transplant (HSCT) context. The focus will be on the agent with the most experience to date, namely the bispecific T-cell engater (BiTE) blinatumomab, but references to antibody-drug conjugates (ADCs) such as inotuzumab ozogamicin and monoclonal antibodies such as daratumamab will be made as well. Specific issues to be addressed include: (1) The use of these agents to reduce measurable residual disease (MRD) prior to HSCT and their potential for improved transplant outcomes due to reduced toxicity compared to traditional chemotherapy salvage, as well as potentially increased toxicity with HSCT with particular agents; (2) the appropriate sequencing of innovative therapies, i.e., when to use BiTEs or antibodies versus CARs pre- and/or post-HSCT; this will include also the potential for impact on response of one group of agents on response to the other; (3) the role of these agents particularly in the post-HSCT relapse setting, or as maintenance to prevent relapse in this setting; (4) special populations in which these agents may substitute for traditional chemotherapy during induction or consolidation in patients with predisposing factors for toxicity with traditional therapy (e.g., Trisomy 21, infants), or those who develop infectious complications precluding delivery of full standard-of-care (SOC) chemotherapy during induction/consolidation (e.g., fungal infections); (5) the evidence we have to date regarding the potential for substitution of blinatumomab for some of the standard chemotherapy agents used pre-HSCT in patients without the above risk factors for toxicity, but with high risk disease going into transplant, in an attempt to decrease current rates of transplant-related mortality as well as morbidity; (6) the unique toxicity profile of these agents and concerns regarding particular side effects in the HSCT context. The manuscript will include both the data we have to date regarding the above issues, ongoing studies that are trying to explore them, and suggestions for future studies to further refine our knowledge base.
Collapse
Affiliation(s)
- Krisztián Miklós Kállay
- Pediatric Hematology and Stem Cell Transplantation Department, National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - Mattia Algeri
- Department of Pediatric Hematology and Oncology, Scientific Institute for Research and Healthcare (IRCCS), Bambino Gesù Childrens' Hospital, Rome, Italy
| | - Jochen Buechner
- Department of Pediatric Hematology and Oncology, Oslo University Hospital, Oslo, Norway
| | - Aviva C. Krauss
- Division of Hematopoietic Stem Cell Transplantation, Department of Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikvah, Israel
| |
Collapse
|
16
|
Hoeben BAW, Wong JYC, Fog LS, Losert C, Filippi AR, Bentzen SM, Balduzzi A, Specht L. Total Body Irradiation in Haematopoietic Stem Cell Transplantation for Paediatric Acute Lymphoblastic Leukaemia: Review of the Literature and Future Directions. Front Pediatr 2021; 9:774348. [PMID: 34926349 PMCID: PMC8678472 DOI: 10.3389/fped.2021.774348] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/03/2021] [Indexed: 12/13/2022] Open
Abstract
Total body irradiation (TBI) has been a pivotal component of the conditioning regimen for allogeneic myeloablative haematopoietic stem cell transplantation (HSCT) in very-high-risk acute lymphoblastic leukaemia (ALL) for decades, especially in children and young adults. The myeloablative conditioning regimen has two aims: (1) to eradicate leukaemic cells, and (2) to prevent rejection of the graft through suppression of the recipient's immune system. Radiotherapy has the advantage of achieving an adequate dose effect in sanctuary sites and in areas with poor blood supply. However, radiotherapy is subject to radiobiological trade-offs between ALL cell destruction, immune and haematopoietic stem cell survival, and various adverse effects in normal tissue. To diminish toxicity, a shift from single-fraction to fractionated TBI has taken place. However, HSCT and TBI are still associated with multiple late sequelae, leaving room for improvement. This review discusses the past developments of TBI and considerations for dose, fractionation and dose-rate, as well as issues regarding TBI setup performance, limitations and possibilities for improvement. TBI is typically delivered using conventional irradiation techniques and centres have locally developed heterogeneous treatment methods and ways to achieve reduced doses in several organs. There are, however, limitations in options to shield organs at risk without compromising the anti-leukaemic and immunosuppressive effects of conventional TBI. Technological improvements in radiotherapy planning and delivery with highly conformal TBI or total marrow irradiation (TMI), and total marrow and lymphoid irradiation (TMLI) have opened the way to investigate the potential reduction of radiotherapy-related toxicities without jeopardising efficacy. The demonstration of the superiority of TBI compared with chemotherapy-only conditioning regimens for event-free and overall survival in the randomised For Omitting Radiation Under Majority age (FORUM) trial in children with high-risk ALL makes exploration of the optimal use of TBI delivery mandatory. Standardisation and comprehensive reporting of conventional TBI techniques as well as cooperation between radiotherapy centres may help to increase the ratio between treatment outcomes and toxicity, and future studies must determine potential added benefit of innovative conformal techniques to ultimately improve quality of life for paediatric ALL patients receiving TBI-conditioned HSCT.
Collapse
Affiliation(s)
- Bianca A. W. Hoeben
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Jeffrey Y. C. Wong
- Department of Radiation Oncology, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, United States
| | - Lotte S. Fog
- Alfred Health Radiation Oncology, The Alfred Hospital, Melbourne, VIC, Australia
| | - Christoph Losert
- Department of Radiation Oncology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Andrea R. Filippi
- Department of Radiation Oncology, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Søren M. Bentzen
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Adriana Balduzzi
- Stem Cell Transplantation Unit, Clinica Paediatrica Università degli Studi di Milano Bicocca, Monza, Italy
| | - Lena Specht
- Department of Oncology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
17
|
The Role of Allogeneic Hematopoietic Stem Cell Transplantation in Pediatric Leukemia. J Clin Med 2021; 10:jcm10173790. [PMID: 34501237 PMCID: PMC8432223 DOI: 10.3390/jcm10173790] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/08/2021] [Accepted: 08/19/2021] [Indexed: 02/07/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) offers potentially curative treatment for many children with high-risk or relapsed acute leukemia (AL), thanks to the combination of intense preparative radio/chemotherapy and the graft-versus-leukemia (GvL) effect. Over the years, progress in high-resolution donor typing, choice of conditioning regimen, graft-versus-host disease (GvHD) prophylaxis and supportive care measures have continuously improved overall transplant outcome, and recent successes using alternative donors have extended the potential application of allotransplantation to most patients. In addition, the importance of minimal residual disease (MRD) before and after transplantation is being increasingly clarified and MRD-directed interventions may be employed to further ameliorate leukemia-free survival after allogeneic HSCT. These advances have occurred in parallel with continuous refinements in chemotherapy protocols and the development of targeted therapies, which may redefine the indications for HSCT in the coming years. This review discusses the role of HSCT in childhood AL by analysing transplant indications in both acute lymphoblastic and acute myeloid leukemia, together with current and most promising strategies to further improve transplant outcome, including optimization of conditioning regimen and MRD-directed interventions.
Collapse
|
18
|
Affiliation(s)
- Nikki A Evensen
- Perlmutter Cancer Center, NYU Grossman School of Medicine New York, New York, NY, USA
| | - William L Carroll
- Perlmutter Cancer Center, NYU Grossman School of Medicine New York, New York, NY, USA.
- Department of Pediatrics and Pathology, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
19
|
Toporski J, Król L, Dykes J, Håkansson Y, Pronk C, Turkiewicz D. The combination of clofarabine, etoposide, and cyclophosphamide shows limited efficacy as a bridge to transplant for children with refractory acute leukemia: results of a monitored prospective study. Pediatr Hematol Oncol 2021; 38:216-226. [PMID: 33150834 DOI: 10.1080/08880018.2020.1838012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Clofarabine has been shown to effectively induce remission in children with refractory leukemia. We conducted a prospective trial (clinicval.trials.gov NCT01025778) to explore the use of clofarabine-based chemotherapy as a bridge-to-transplant approach. Children with refractory acute leukemia were enrolled to receive two induction courses of clofarabine, etoposide, and cyclophosphamide (CloEC). Responding patients were scheduled for T-cell depleted haploidentical hematopoietic stem cell transplantation (HSCT). The primary objective was to improve survival by achieving sufficient disease control to enable stem cell transplantation. Secondary objectives were to evaluate safety and toxicity. Seven children with active disease entered the study. Two children responded to induction courses and underwent transplantation. Five children did not respond to induction: one died in progression after the first course; two received off-protocol chemotherapy and were transplanted; and two succumbed to progressive leukemia. All transplanted children engrafted and no acute skin graft-versus-host disease > grade I was observed. One child is alive and well 7.5 years after the first CloEC course. One child developed fulminant adenovirus hepatitis and died in continuous complete remission 7 months after start of induction. Two children relapsed and died 6.5 and 7.5 months after enrollment. Infection was the most common toxicity. CloEC can induce responses in some patients with refractory acute leukemia but is highly immunosuppressive, resulting in substantial risk of life-threatening infections. In our study, haploidentical HSCT was feasible with sustained engraftment. No clinically significant organ toxicity was observed. Also, repeating CloEC probably does not increase the chance of achieving remission.
Collapse
Affiliation(s)
- Jacek Toporski
- Department of Pediatrics, Section of Pediatric Oncology, Hematology, Immunology and Nephrology, Skåne University Hospital, Lund, Sweden
| | - Ladislav Król
- Department of Pediatrics, Section of Pediatric Oncology, Hematology, Immunology and Nephrology, Skåne University Hospital, Lund, Sweden
| | - Josefina Dykes
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, University and Regional Laboratories, Lund, Sweden
| | - Yvonne Håkansson
- Department of Pediatrics, Section of Pediatric Oncology, Hematology, Immunology and Nephrology, Skåne University Hospital, Lund, Sweden
| | - Cornelis Pronk
- Department of Pediatrics, Section of Pediatric Oncology, Hematology, Immunology and Nephrology, Skåne University Hospital, Lund, Sweden
| | - Dominik Turkiewicz
- Department of Pediatrics, Section of Pediatric Oncology, Hematology, Immunology and Nephrology, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
20
|
Cao LQ, Zhou Y, Liu YR, Xu LP, Zhang XH, Wang Y, Chen H, Chen YH, Wang FR, Han W, Sun YQ, Yan CH, Tang FF, Mo XD, Liu KY, Fan QZ, Chang YJ, Huang XJ. A risk score system for stratifying the risk of relapse in B cell acute lymphocytic leukemia patients after allogenic stem cell transplantation. Chin Med J (Engl) 2021; 134:1199-1208. [PMID: 33734137 PMCID: PMC8143760 DOI: 10.1097/cm9.0000000000001402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND For patients with B cell acute lymphocytic leukemia (B-ALL) who underwent allogeneic stem cell transplantation (allo-SCT), many variables have been demonstrated to be associated with leukemia relapse. In this study, we attempted to establish a risk score system to predict transplant outcomes more precisely in patients with B-ALL after allo-SCT. METHODS A total of 477 patients with B-ALL who underwent allo-SCT at Peking University People's Hospital from December 2010 to December 2015 were enrolled in this retrospective study. We aimed to evaluate the factors associated with transplant outcomes after allo-SCT, and establish a risk score to identify patients with different probabilities of relapse. The univariate and multivariate analyses were performed with the Cox proportional hazards model with time-dependent variables. RESULTS All patients achieved neutrophil engraftment, and 95.4% of patients achieved platelet engraftment. The 5-year cumulative incidence of relapse (CIR), overall survival (OS), leukemia-free survival (LFS), and non-relapse mortality were 20.7%, 70.4%, 65.6%, and 13.9%, respectively. Multivariate analysis showed that patients with positive post-transplantation minimal residual disease (MRD), transplanted beyond the first complete remission (≥CR2), and without chronic graft-versus-host disease (cGVHD) had higher CIR (P < 0.001, P = 0.004, and P < 0.001, respectively) and worse LFS (P < 0.001, P = 0.017, and P < 0.001, respectively), and OS (P < 0.001, P = 0.009, and P < 0.001, respectively) than patients without MRD after transplantation, transplanted in CR1, and with cGVHD. A risk score for predicting relapse was formulated with the three above variables. The 5-year relapse rates were 6.3%, 16.6%, 55.9%, and 81.8% for patients with scores of 0, 1, 2, and 3 (P < 0.001), respectively, while the 5-year LFS and OS values decreased with increasing risk score. CONCLUSION This new risk score system might stratify patients with different risks of relapse, which could guide treatment.
Collapse
Affiliation(s)
- Le-Qing Cao
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Yang Zhou
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Yan-Rong Liu
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Lan-Ping Xu
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Yu Wang
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Huan Chen
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Yu-Hong Chen
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Feng-Rong Wang
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Wei Han
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Yu-Qian Sun
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Chen-Hua Yan
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Fei-Fei Tang
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Xiao-Dong Mo
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Kai-Yan Liu
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Qiao-Zhen Fan
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Ying-Jun Chang
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Xiao-Jun Huang
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, 2019RU029, Beijing, China
| |
Collapse
|
21
|
Ruggeri A, Galimard JE, Paina O, Fagioli F, Tbakhi A, Yesilipek A, Navarro JMF, Faraci M, Hamladji RM, Skorobogatova E, Al-Seraihy A, Sundin M, Herrera C, Rifón J, Dalissier A, Locatelli F, Rocha V, Corbacioglu S. Outcomes of Unmanipulated Haploidentical Transplantation Using Post-Transplant Cyclophosphamide (PT-Cy) in Pediatric Patients With Acute Lymphoblastic Leukemia. Transplant Cell Ther 2021; 27:424.e1-424.e9. [PMID: 33965182 DOI: 10.1016/j.jtct.2021.01.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 01/14/2021] [Accepted: 01/18/2021] [Indexed: 12/18/2022]
Abstract
HLA-haploidentical transplantation (haplo-HCT) using post-transplantation-cyclophosphamide (PT-Cy) is a feasible procedure in children with malignancies. However, large studies on Haplo-HCT with PT-Cy for childhood acute lymphoblastic leukemia (ALL) are lacking. We analyzed haplo-HCT outcomes in 180 children with ALL. Median age was 9 years, and median follow-up was 2.7 years. Disease status was CR1 for 24%, CR2 for 45%, CR+3 for 12%, and active disease for 19%. All patients received PT-Cy day +3 and +4. Bone marrow (BM) was the stem cell source in 115 patients (64%). Cumulative incidence of 42-day engraftment was 88.9%. Cumulative incidence of day-100 acute graft-versus-host disease (GVHD) grade II-IV was 28%, and 2-year chronic GVHD was 21.9%. At 2 years, cumulative incidence of nonrelapse mortality (NRM) was 19.6%. Cumulative incidence was 41.9% for relapse and 25% for patients in CR1. Estimated 2-year leukemia free survival was 65%, 44%, and 18.8% for patients transplanted in CR1, CR2, CR3+ and 3% at 1 year for active disease. In multivariable analysis for patients in CR1 and CR2, disease status (CR2 [hazard ratio {HR} = 2.19; P = .04]), age at HCT older than 13 (HR = 2.07; P = .03) and use of peripheral blood stem cell (PBSC) (HR = 1.98; P = .04) were independent factors associated with decreased overall survival. Use of PBSC was also associated with higher NRM (HR = 3.13; P = .04). Haplo-HCT with PT-Cy is an option for children with ALL, namely those transplanted in CR1 and CR2. Age and disease status remain the most important factors for outcomes. BM cells as a graft source is associated with improved survival.
Collapse
Affiliation(s)
- Annalisa Ruggeri
- Department of Hematology and Bone marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milano, Italy; Cellular Therapy and Immunobiology Working Party, Leiden, The Netherlands.
| | | | - Olesya Paina
- First State Pavlov Medical University of St. Petersburg Raisa Gorbacheva Memorial Research Institute for Paediatric Oncology, Hematology and Transplantation, St. Petersburg, Russia
| | - Franca Fagioli
- Onco-Ematologia Pediatrica, Centro Trapianti Cellule Staminali, Ospedale Infantile Regina Margherita, Torino, Italy
| | | | | | | | | | | | - Elena Skorobogatova
- The Russian Children´s Research Hospital, Department of Bone Marrow Transplantation, Moscow, Russia
| | - Amal Al-Seraihy
- King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Mikael Sundin
- Pediatric Hematology, Immunology and HCT Section, Astrid Lindgren Children's Hospital, Karolinska University Hospital and Division of Pedatrics, CLINTEC, Karolinska Institutet; Stockholm, Sweden
| | - Concepcion Herrera
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Department of Hematology Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Jose Rifón
- Clínica Universitaria de Navarra Area de Terápia Celular Pamplona, Pamplona, Spain
| | | | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Vanderson Rocha
- Service of Hematology, Transfusion and Cell Therapy and Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology, HCFMUSP, University of São Paulo Medical School, São Paulo, Brazil
| | - Selim Corbacioglu
- Pediatric Hematology, Oncology and Stem Cell Transplantation Department, University of Regensburg, Regensburg, Germany
| |
Collapse
|
22
|
Merli P, Ifversen M, Truong TH, Marquart HV, Buechner J, Wölfl M, Bader P. Minimal Residual Disease Prior to and After Haematopoietic Stem Cell Transplantation in Children and Adolescents With Acute Lymphoblastic Leukaemia: What Level of Negativity Is Relevant? Front Pediatr 2021; 9:777108. [PMID: 34805054 PMCID: PMC8602790 DOI: 10.3389/fped.2021.777108] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/14/2021] [Indexed: 12/18/2022] Open
Abstract
Minimal residual disease (MRD) assessment plays a central role in risk stratification and treatment guidance in paediatric patients with acute lymphoblastic leukaemia (ALL). As such, MRD prior to haematopoietic stem cell transplantation (HSCT) is a major factor that is independently correlated with outcome. High burden of MRD is negatively correlated with post-transplant survival, as both the risk of leukaemia recurrence and non-relapse mortality increase with greater levels of MRD. Despite growing evidence supporting these findings, controversies still exist. In particular, it is still not clear whether multiparameter flow cytometry and real-time quantitative polymerase chain reaction, which is used to recognise immunoglobulin and T-cell receptor gene rearrangements, can be employed interchangeably. Moreover, the higher sensitivity in MRD quantification offered by next-generation sequencing techniques may further refine the ability to stratify transplant-associated risks. While MRD quantification from bone marrow prior to HSCT remains the state of the art, heavily pre-treated patients may benefit from additional staging, such as using 18F-fluorodeoxyglucose positron emission tomography/computed tomography to detect focal residues of disease. Additionally, the timing of MRD detection (i.e., immediately before administration of the conditioning regimen or weeks before) is a matter of debate. Pre-transplant MRD negativity has previously been associated with superior outcomes; however, in the recent For Omitting Radiation Under Majority age (FORUM) study, pre-HSCT MRD positivity was associated with neither relapse risk nor survival. In this review, we discuss the level of MRD that may require pre-transplant therapy intensification, risking time delay and complications (as well as losing the window for HSCT if disease progression occurs), as opposed to an adapted post-transplant strategy to achieve long-term remission. Indeed, MRD monitoring may be a valuable tool to guide individualised treatment decisions, including tapering of immunosuppression, cellular therapies (such as donor lymphocyte infusions) or additional immunotherapy (such as bispecific T-cell engagers or chimeric antigen receptor T-cell therapy).
Collapse
Affiliation(s)
- Pietro Merli
- Department of Pediatric Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Marianne Ifversen
- Pediatric Stem Cell Transplant and Immune Deficiency, Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Tony H Truong
- Division of Pediatric Oncology and Bone Marrow Transplant, Alberta Children's Hospital, University of Calgary, Calgary, AB, Canada
| | - Hanne V Marquart
- Section for Diagnostic Immunology, Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jochen Buechner
- Department of Pediatric Hematology and Oncology, Oslo University Hospital, Oslo, Norway
| | - Matthias Wölfl
- Pediatric Hematology, Oncology and Stem Cell Transplantation, Children's Hospital, Würzburg University Hospital, Würzburg, Germany
| | - Peter Bader
- Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Department for Children and Adolescents, Goethe University, University Hospital Frankfurt, Frankfurt, Germany
| |
Collapse
|
23
|
Lew G, Chen Y, Lu X, Rheingold SR, Whitlock JA, Devidas M, Hastings CA, Winick NJ, Carroll WL, Wood BL, Borowitz MJ, Pulsipher MA, Hunger SP. Outcomes after late bone marrow and very early central nervous system relapse of childhood B-acute lymphoblastic leukemia: a report from the Children's Oncology Group phase III study AALL0433. Haematologica 2021; 106:46-55. [PMID: 32001530 PMCID: PMC7776266 DOI: 10.3324/haematol.2019.237230] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/24/2020] [Indexed: 12/18/2022] Open
Abstract
Outcomes after relapse of childhood B-acute lymphoblastic leukemia (B-ALL) are poor, and optimal therapy is unclear. The children’s Oncology Group study AALL0433 evaluated a new platform for relapsed ALL. Between March 2007 and October 2013 AALL0433 enrolled 275 participants with late bone marrow or very early isolated central nervous system (iCNS) relapse of childhood B-ALL. Patients were randomized to receive standard versus intensive vincristine dosing; this randomization was closed due to excess peripheral neuropathy in 2010. Patients with matched sibling donors received allogeneic hematopoietic cell transplantation (HCT) after the first three blocks of therapy. The prognostic value of minimal residual disease (MRD) was also evaluated in this study. The 3-year event free and overall survival (EFS/OS) for the 271 eligible patients were 63.6±3.0% and 72.3±2.8% respectively. MRD at the end of Induction-1 was highly predictive of outcome, with 3-year EFS/OS of 84.9±4.0% and 93.8±2.7% for patients with MRD <0.1%, versus 53.7±7.8% and 60.6± 7.8% for patients with MRD ≥0.1% (P<0.0001). Patients who received HCT versus chemotherapy alone had an improved 3-year disease-free survival (77.5±6.2% vs. 66.9 + 4.5%, P=0.03) but not OS (81.5±5.8% for HCT vs. 85.8±3.4% for chemotherapy, P=0.46). Patients with early iCNS relapse fared poorly, with a 3-year EFS/OS of 41.4±9.2% and 51.7±9.3%, respectively. Infectious toxicities of the chemotherapy platform were significant. The AALL0433 chemotherapy platform is efficacious for late bone marrow relapse of B-ALL, but with significant toxicities. The MRD threshold of 0.1% at the end of Induction-1 was highly predictive of the outcome. The optimal role for HCT for this patient population remains uncertain. This trial is registered at clinicaltrials.gov (NCT# 00381680).
Collapse
Affiliation(s)
- Glen Lew
- Emory University / Children's Healthcare of Atlanta
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Wölfl M, Qayed M, Benitez Carabante MI, Sykora T, Bonig H, Lawitschka A, Diaz-de-Heredia C. Current Prophylaxis and Treatment Approaches for Acute Graft-Versus-Host Disease in Haematopoietic Stem Cell Transplantation for Children With Acute Lymphoblastic Leukaemia. Front Pediatr 2021; 9:784377. [PMID: 35071133 PMCID: PMC8771910 DOI: 10.3389/fped.2021.784377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/09/2021] [Indexed: 11/13/2022] Open
Abstract
Acute graft-versus-host disease (aGvHD) continues to be a leading cause of morbidity and mortality following allogeneic haematopoietic stem cell transplantation (HSCT). However, higher event-free survival (EFS) was observed in patients with acute lymphoblastic leukaemia (ALL) and grade II aGvHD vs. patients with no or grade I GvHD in the randomised, controlled, open-label, international, multicentre Phase III For Omitting Radiation Under Majority age (FORUM) trial. This finding suggests that moderate-severity aGvHD is associated with a graft-versus-leukaemia effect which protects against leukaemia recurrence. In order to optimise the benefits of HSCT for leukaemia patients, reduction of non-relapse mortality-which is predominantly caused by severe GvHD-is of utmost importance. Herein, we review contemporary prophylaxis and treatment options for aGvHD in children with ALL and the key challenges of aGvHD management, focusing on maintaining the graft-versus-leukaemia effect without increasing the severity of GvHD.
Collapse
Affiliation(s)
- Matthias Wölfl
- Pediatric Hematology, Oncology and Stem Cell Transplantation, Children's Hospital, Würzburg University Hospital, Würzburg, Germany
| | - Muna Qayed
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, Atlanta, GA, United States
| | - Maria Isabel Benitez Carabante
- Department of Pediatric Hematology and Oncology, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Tomas Sykora
- Haematopoietic Stem Cell Transplantation Unit, Department of Pediatric Haematology and Oncology, Comenius University Children's Hospital, Bratislava, Slovakia
| | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohematology, Goethe-University Frankfurt/Main, Frankfurt, Germany.,German Red Cross Blood Service BaWüHe, Frankfurt, Germany
| | - Anita Lawitschka
- Department of Pediatrics, St. Anna Kinderspital and Children's Cancer Research Institute, Medical University of Vienna, Vienna, Austria
| | - Cristina Diaz-de-Heredia
- Department of Pediatric Hematology and Oncology, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| |
Collapse
|
25
|
Diorio C, Maude SL. CAR T cells vs allogeneic HSCT for poor-risk ALL. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2020; 2020:501-507. [PMID: 33275706 PMCID: PMC7727575 DOI: 10.1182/hematology.2020000172] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
For subgroups of children with B-cell acute lymphoblastic leukemia (B-ALL) at very high risk of relapse, intensive multiagent chemotherapy has failed. Traditionally, the field has turned to allogeneic hematopoietic stem cell transplantation (HSCT) for patients with poor outcomes. While HSCT confers a survival benefit for several B-ALL populations, often HSCT becomes standard-of-care in subsets of de novo ALL with poor risk features despite limited or no data showing a survival benefit in these populations, yet the additive morbidity and mortality can be substantial. With the advent of targeted immunotherapies and the transformative impact of CD19-directed chimeric antigen receptor (CAR)-modified T cells on relapsed or refractory B-ALL, this approach is currently under investigation in frontline therapy for a subset of patients with poor-risk B-ALL: high-risk B-ALL with persistent minimal residual disease at the end of consolidation, which has been designated very high risk. Comparisons of these 2 approaches are fraught with issues, including single-arm trials, differing eligibility criteria, comparisons to historical control populations, and vastly different toxicity profiles. Nevertheless, much can be learned from available data and ongoing trials. We will review data for HSCT for pediatric B-ALL in first remission and the efficacy of CD19 CAR T-cell therapy in relapsed or refractory B-ALL, and we will discuss an ongoing international phase 2 clinical trial of CD19 CAR T cells for very-high-risk B-ALL in first remission.
Collapse
Affiliation(s)
- Caroline Diorio
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA; and
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Shannon L. Maude
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA; and
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA
| |
Collapse
|
26
|
Scarpelli DB, Murphy B, Chang BH, Nemecek E, Jaboin JJ. In Regard to Wong et al. Int J Radiat Oncol Biol Phys 2020; 108:1395-1396. [PMID: 33220230 DOI: 10.1016/j.ijrobp.2020.07.2316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 07/23/2020] [Indexed: 10/23/2022]
Affiliation(s)
- Daphne B Scarpelli
- Department of Radiation Medicine, Oregon Health & Science University, Portland, Oregon
| | - Blair Murphy
- Department of Radiation Medicine, Oregon Health & Science University, Portland, Oregon
| | - Bill H Chang
- Department of Pediatrics, Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Oregon Health & Science University, Portland, Oregon
| | - Eneida Nemecek
- Department of Pediatrics, Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Oregon Health & Science University, Portland, Oregon
| | - Jerry J Jaboin
- Department of Radiation Medicine, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
27
|
Saint Fleur-Lominy S, Evensen NA, Bhatla T, Sethia G, Narang S, Choi JH, Ma X, Yang JJ, Kelly S, Raetz E, Harvey RC, Willman C, Loh ML, Hunger SP, Brown PA, Getz KM, Meydan C, Mason CE, Tsirigos A, Carroll WL. Evolution of the Epigenetic Landscape in Childhood B Acute Lymphoblastic Leukemia and Its Role in Drug Resistance. Cancer Res 2020; 80:5189-5202. [PMID: 33067268 DOI: 10.1158/0008-5472.can-20-1145] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 08/13/2020] [Accepted: 10/12/2020] [Indexed: 11/16/2022]
Abstract
Although B-cell acute lymphoblastic leukemia (B-ALL) is the most common malignancy in children and while highly curable, it remains a leading cause of cancer-related mortality. The outgrowth of tumor subclones carrying mutations in genes responsible for resistance to therapy has led to a Darwinian model of clonal selection. Previous work has indicated that alterations in the epigenome might contribute to clonal selection, yet the extent to which the chromatin state is altered under the selective pressures of therapy is unknown. To address this, we performed chromatin immunoprecipitation, gene expression analysis, and enhanced reduced representation bisulfite sequencing on a cohort of paired diagnosis and relapse samples from individual patients who all but one relapsed within 36 months of initial diagnosis. The chromatin state at diagnosis varied widely among patients, while the majority of peaks remained stable between diagnosis and relapse. Yet a significant fraction was either lost or newly gained, with some patients showing few differences and others showing massive changes of the epigenetic state. Evolution of the epigenome was associated with pathways previously linked to therapy resistance as well as novel candidate pathways through alterations in pyrimidine biosynthesis and downregulation of polycomb repressive complex 2 targets. Three novel, relapse-specific superenhancers were shared by a majority of patients including one associated with S100A8, the top upregulated gene seen at relapse in childhood B-ALL. Overall, our results support a role of the epigenome in clonal evolution and uncover new candidate pathways associated with relapse. SIGNIFICANCE: This study suggests a major role for epigenetic mechanisms in driving clonal evolution in B-ALL and identifies novel pathways associated with drug resistance.
Collapse
Affiliation(s)
- Shella Saint Fleur-Lominy
- Perlmutter Cancer Center, NYU Langone Health, New York, New York.,Department of Medicine, NYU Langone Health, New York, New York
| | - Nikki A Evensen
- Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Teena Bhatla
- Department of Pediatrics, Children's Hospital of New Jersey at NBI, RWJBarnabas Health, Newark, New Jersey
| | - Gunjan Sethia
- Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Sonali Narang
- Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Jun H Choi
- Department of Medicine, NYU Langone Health, New York, New York
| | - Xiaotu Ma
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jun J Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Stephen Kelly
- Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Elizabeth Raetz
- Perlmutter Cancer Center, NYU Langone Health, New York, New York.,Department of Pediatrics, NYU Health, New York, New York
| | - Richard C Harvey
- University of New Mexico Comprehensive Cancer Center, Department of Pathology, University of New Mexico School of Medicine and Health Sciences Center, Albuquerque, New Mexico
| | - Cheryl Willman
- University of New Mexico Comprehensive Cancer Center, Department of Pathology, University of New Mexico School of Medicine and Health Sciences Center, Albuquerque, New Mexico
| | - Mignon L Loh
- Department of Pediatrics, UCSF Benioff Children's Hospital, San Francisco, California
| | - Stephen P Hunger
- Department of Pediatrics and the Center for Childhood Cancer Research, Children's Hospital of Philadelphia and The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Patrick A Brown
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kylie M Getz
- Department of Physiology and Biophysics and Institute for Computational Biomedicine and Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York
| | - Cem Meydan
- Department of Physiology and Biophysics and Institute for Computational Biomedicine and Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York
| | - Christopher E Mason
- Department of Physiology and Biophysics and Institute for Computational Biomedicine and Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York
| | - Aristotelis Tsirigos
- Perlmutter Cancer Center, NYU Langone Health, New York, New York. .,Department of Pathology, NYU Langone Health, New York, New York
| | - William L Carroll
- Perlmutter Cancer Center, NYU Langone Health, New York, New York. .,Department of Pediatrics, NYU Health, New York, New York.,Department of Pathology, NYU Langone Health, New York, New York
| |
Collapse
|
28
|
Gupta S, Maude SL, O'Brien MM, Rau RE, McNeer JL. How the COG is Approaching the High-Risk Patient with ALL: Incorporation of Immunotherapy into Frontline Treatment. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2020; 20 Suppl 1:S8-S11. [PMID: 32862880 DOI: 10.1016/s2152-2650(20)30443-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Affiliation(s)
- Sumit Gupta
- Hospital for Sick Children and University of Toronto, 555 University Ave, Toronto, ON, M5G 1X8, Canada
| | - Shannon L Maude
- Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, 3501 Civic Center Blvd, Philadelphia, PA, 19104, United States
| | - Maureen M O'Brien
- Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Ave, Cincinnati, OH, 45229, United States
| | - Rachel E Rau
- Baylor College of Medicine, Texas Children's Hospital, 1102 Bates Street, Houston, TX, 77030, United States
| | - Jennifer L McNeer
- University of Chicago Comer Children's Hospital, 5841 S Maryland Ave, Chicago, IL, 60637, United States.
| |
Collapse
|
29
|
Merli P, Algeri M, Del Bufalo F, Locatelli F. Hematopoietic Stem Cell Transplantation in Pediatric Acute Lymphoblastic Leukemia. Curr Hematol Malig Rep 2020; 14:94-105. [PMID: 30806963 DOI: 10.1007/s11899-019-00502-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE OF REVIEW The remarkable improvement in the prognosis of children with acute lymphoblastic leukemia (ALL) has been mainly achieved through the administration of risk-adapted therapy, including allogeneic hematopoietic stem cell transplantation (HSCT). This paper reviews the current indications to HSCT in ALL children, as well as the type of donor and conditioning regimens commonly used. Finally, it will focus on future challenges in immunotherapy. RECENT FINDINGS As our comprehension of disease-specific risk factors improves, indications to HSCT continue to evolve. Future studies will answer the year-old question on the best conditioning regimen to be used in this setting, while a recent randomized controlled study fixed the optimal anti-thymocyte globulin dose in unrelated donor HSCT. HSCT, the oldest immunotherapy used in clinical practice, still represents the gold standard consolidation treatment for a number of pediatric patients with high-risk/relapsed ALL. New immunotherapies hold the promise of further improving outcomes in this setting.
Collapse
Affiliation(s)
- Pietro Merli
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, Piazza Sant'Onofrio, 4, 00165, Rome, Italy
| | - Mattia Algeri
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, Piazza Sant'Onofrio, 4, 00165, Rome, Italy
| | - Francesca Del Bufalo
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, Piazza Sant'Onofrio, 4, 00165, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, Piazza Sant'Onofrio, 4, 00165, Rome, Italy. .,Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
30
|
McNeer JL, Rau RE, Gupta S, Maude SL, O'Brien MM. Cutting to the Front of the Line: Immunotherapy for Childhood Acute Lymphoblastic Leukemia. Am Soc Clin Oncol Educ Book 2020; 40:1-12. [PMID: 32320280 DOI: 10.1200/edbk_278171] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Although many children and young adults with B-cell acute lymphoblastic leukemia (B-ALL) are cured with modern, risk-adapted chemotherapy regimens, 10% to 15% of patients will experience relapse or have refractory disease. Recent efforts to further intensify cytotoxic chemotherapy regimens in the frontline setting have failed as a result of excessive toxicity or lack of improvement in efficacy. As a result, novel approaches will be required to achieve cures in more newly diagnosed patients. Multiple immune-based therapies have demonstrated considerable efficacy in the setting of relapsed or refractory (R/R) disease, including CD19 targeting with blinatumomab and tisagenlecleucel and CD22 targeting with inotuzumab ozogamicin. These agents are now under investigation by the Children's Oncology Group (COG) in clinical trials for newly diagnosed B-ALL, with integration into standard chemotherapy regimens based on clinically and biology-based risk stratification as well as disease response.
Collapse
Affiliation(s)
| | - Rachel E Rau
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX
| | - Sumit Gupta
- The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Shannon L Maude
- Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Maureen M O'Brien
- Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
31
|
Bader P, Salzmann-Manrique E, Balduzzi A, Dalle JH, Woolfrey AE, Bar M, Verneris MR, Borowitz MJ, Shah NN, Gossai N, Shaw PJ, Chen AR, Schultz KR, Kreyenberg H, Di Maio L, Cazzaniga G, Eckert C, van der Velden VHJ, Sutton R, Lankester A, Peters C, Klingebiel TE, Willasch AM, Grupp SA, Pulsipher MA. More precisely defining risk peri-HCT in pediatric ALL: pre- vs post-MRD measures, serial positivity, and risk modeling. Blood Adv 2019; 3:3393-3405. [PMID: 31714961 PMCID: PMC6855112 DOI: 10.1182/bloodadvances.2019000449] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/14/2019] [Indexed: 12/14/2022] Open
Abstract
Detection of minimal residual disease (MRD) pre- and post-hematopoietic cell transplantation (HCT) for pediatric acute lymphoblastic leukemia (ALL) has been associated with relapse and poor survival. Published studies have had insufficient numbers to: (1) compare the prognostic value of pre-HCT and post-HCT MRD; (2) determine clinical factors post-HCT associated with better outcomes in MRD+ patients; and (3) use MRD and other clinical factors to develop and validate a prognostic model for relapse in pediatric patients with ALL who undergo allogeneic HCT. To address these issues, we assembled an international database including sibling (n = 191), unrelated (n = 259), mismatched (n = 56), and cord blood (n = 110) grafts given after myeloablative conditioning. Although high and very high MRD pre-HCT were significant predictors in univariate analysis, with bivariate analysis using MRD pre-HCT and post-HCT, MRD pre-HCT at any level was less predictive than even low-level MRD post-HCT. Patients with MRD pre-HCT must become MRD low/negative at 1 to 2 months and negative within 3 to 6 months after HCT for successful therapy. Factors associated with improved outcome of patients with detectable MRD post-HCT included acute graft-versus-host disease. We derived a risk score with an MRD cohort from Europe, North America, and Australia using negative predictive characteristics (late disease status, non-total body irradiation regimen, and MRD [high, very high]) defining good, intermediate, and poor risk groups with 2-year cumulative incidences of relapse of 21%, 38%, and 47%, respectively. We validated the score in a second, more contemporaneous cohort and noted 2-year cumulative incidences of relapse of 13%, 26%, and 47% (P < .001) for the defined risk groups.
Collapse
Affiliation(s)
- Peter Bader
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Emilia Salzmann-Manrique
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Adriana Balduzzi
- Clinica Pediatrica, Università degli Studi di Milano-Bicocca, Ospedale San Gerardo, Monza, Italy
| | - Jean-Hugues Dalle
- Department of Pediatric Hemato-Immunology, Hôpital Robert Debré and Paris-Diderot University, Paris, France
| | - Ann E Woolfrey
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA
| | - Merav Bar
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA
| | - Michael R Verneris
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Michael J Borowitz
- Department of Pathology, John Hopkins Medical Institutions, Baltimore, MD
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, MD
| | - Nathan Gossai
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Peter J Shaw
- BMT Services, Sydney Children's Hospital Network, Westmead, Sydney, NSW, Australia
| | - Allen R Chen
- Pediatric Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD
| | - Kirk R Schultz
- Division of Hematology/Oncology/Bone Marrow Transplantation, Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Hermann Kreyenberg
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Lucia Di Maio
- Clinica Pediatrica, Università degli Studi di Milano-Bicocca, Ospedale San Gerardo, Monza, Italy
| | - Gianni Cazzaniga
- Clinica Pediatrica, Università degli Studi di Milano-Bicocca, Ospedale San Gerardo, Monza, Italy
| | - Cornelia Eckert
- Charité University Medical Center Berlin, Children's Hospital, Berlin, Germany
| | | | - Rosemary Sutton
- School of Women's and Children's Health, Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Randwick, NSW, Australia
| | - Arjan Lankester
- Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Christina Peters
- St Anna Children's Hospital, Universitätsklinik für Kinder und Jugendheilkunde, Medizinische Universität Wien, Vienna, Austria
| | - Thomas E Klingebiel
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Andre M Willasch
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Stephan A Grupp
- Pediatric Oncology, The Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; and
| | - Michael A Pulsipher
- Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, CA
| |
Collapse
|
32
|
Balduzzi A, Dalle JH, Wachowiak J, Yaniv I, Yesilipek A, Sedlacek P, Bierings M, Ifversen M, Sufliarska S, Kalwak K, Lankester A, Toporski J, Di Maio L, Glogova E, Poetschger U, Peters C. Transplantation in Children and Adolescents with Acute Lymphoblastic Leukemia from a Matched Donor versus an HLA-Identical Sibling: Is the Outcome Comparable? Results from the International BFM ALL SCT 2007 Study. Biol Blood Marrow Transplant 2019; 25:2197-2210. [PMID: 31319153 DOI: 10.1016/j.bbmt.2019.07.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 07/07/2019] [Accepted: 07/08/2019] [Indexed: 12/19/2022]
Abstract
Eligibility criteria for hematopoietic stem cell transplantation (HSCT) in acute lymphoblastic leukemia (ALL) vary according to disease characteristics, response to treatment, and type of available donor. As the risk profile of the patient worsens, a wider degree of HLA mismatching is considered acceptable. A total of 138 children and adolescents who underwent HSCT from HLA-identical sibling donors (MSDs) and 210 who underwent HSCT from matched donors (MDs) (median age, 9 years; 68% male) in 10 countries were enrolled in the International-BFM ALL SCT 2007 prospective study to assess the impact of donor type in HSCT for pediatric ALL. The 4-year event-free survival (65 ± 5% vs 61 ± 4%; P = .287), overall survival (72 ± 4% versus 68 ± 4%; P = .235), cumulative incidence of relapse (24 ± 4% versus 25 ± 3%; P = .658) and nonrelapse mortality (10 ± 3% versus 14 ± 3%; P = .212) were not significantly different between MSD and MD graft recipients. The risk of extensive chronic (cGVHD) was lower in MD graft recipients than in MSD graft recipients (hazard ratio [HR], .38; P = .002), and the risks of severe acute GVHD (aGVHD) and cGVHD were higher in peripheral blood stem cell graft recipients than in bone marrow graft recipients (HR, 2.06; P = .026). Compared with the absence of aGVHD, grade I-II aGVHD was associated with a lower risk of graft failure (HR, .63; P = .042) and grade III-IV aGVHD was associated with a higher risk of graft failure (HR, 1.85; P = .020) and nonleukemic death (HR, 8.76; P < .0001), despite a lower risk of relapse (HR, .32; P = .021). Compared with the absence of cGVHD, extensive cGVHD was associated with a higher risk of nonleukemic death (HR, 8.12; P < .0001). Because the outcomes of transplantation from a matched donor were not inferior to those of transplantation from an HLA-identical sibling, eligibility criteria for transplantation might be reviewed in pediatric ALL and possibly in other malignancies as well. Bone marrow should be the preferred stem cell source, and the addition of MTX should be considered in MSD graft recipients.
Collapse
Affiliation(s)
- Adriana Balduzzi
- Clinica Pediatrica, Università degli Studi di Milano Bicocca, Fondazione Monza e Brianza per il Bambino e la sua Mamma, Ospedale San Gerardo, Monza, Italy.
| | - Jean-Hugues Dalle
- Hemato-Immunology Department, Robert-Debre Hospital, APHP and Paris-Diderot University, Paris, France
| | - Jacek Wachowiak
- Department of Pediatric Oncology, Hematology and Transplantology, University of Medical Sciences, Poznan, Poland
| | - Isaac Yaniv
- Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Akif Yesilipek
- Antalya Medicalpark Hospital, Pediatric Stem Cell Transplantation Unit, Antalya, Turkey
| | - Petr Sedlacek
- Department of Pediatric Hematology and Oncology, University Hospital Motol, Prague, Czech Republic
| | - Marc Bierings
- Princess Maxima Centre for Pediatric Oncology and Utrecht University Children's Hospital, Utrecht, The Netherlands
| | - Marianne Ifversen
- Department of Pediatric and Adolescent Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sabina Sufliarska
- Bone Marrow Transplantation Unit, Comenius University Children's Hospital, Bratislava, Slovakia, Bratislava, Slovakia
| | - Krzysztof Kalwak
- Department of Pediatric Hematology/Oncology and BMT, Wroclaw Medical University, Wroclaw, Poland
| | - Arjan Lankester
- Department of Pediatrics, University Medical Centre, Willem-Alexander Children's Hospital, Leiden, The Netherlands
| | - Jacek Toporski
- Children's Hospital, Skåne University Hospital, Lund, Sweden
| | - Lucia Di Maio
- Clinica Pediatrica, Università degli Studi di Milano Bicocca, Fondazione Monza e Brianza per il Bambino e la sua Mamma, Ospedale San Gerardo, Monza, Italy
| | | | | | | |
Collapse
|
33
|
Esiashvili N, Lu X, Ulin K, Laurie F, Kessel S, Kalapurakal JA, Merchant TE, Followill DS, Sathiaseelan V, Schmitter MK, Devidas M, Chen Y, Wall DA, Brown PA, Hunger SP, Grupp SA, Pulsipher MA. Higher Reported Lung Dose Received During Total Body Irradiation for Allogeneic Hematopoietic Stem Cell Transplantation in Children With Acute Lymphoblastic Leukemia Is Associated With Inferior Survival: A Report from the Children's Oncology Group. Int J Radiat Oncol Biol Phys 2019; 104:513-521. [PMID: 30807822 PMCID: PMC6548591 DOI: 10.1016/j.ijrobp.2019.02.034] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 01/14/2019] [Accepted: 02/14/2019] [Indexed: 12/11/2022]
Abstract
PURPOSE To examine the relationship between lung radiation dose and survival outcomes in children undergoing total body irradiation (TBI)-based hematopoietic stem cell transplantation (HSCT) for acute lymphoblastic leukemia on the Children's Oncology Group trial. METHODS AND MATERIALS TBI (1200 or 1320 cGy given twice daily in 6 or 8 fractions) was used as part of 3 HSCT preparative regimens, allowing institutional flexibility regarding TBI techniques, including lung shielding. Lung doses as reported by each participating institution were calculated for different patient setups, with and without shielding, with a variety of dose calculation techniques. The association between lung dose and transplant-related mortality, relapse-free survival, and overall survival (OS) was examined using the Cox proportional hazards regression model controlling for the following variables: TBI dose rate, TBI fields, patient position during TBI, donor type, and pre-HSCT minimal residual disease level. RESULTS Of a total of 143 eligible patients, 127 had lung doses available for this analysis. The TBI techniques were heterogeneous. The mean lung dose was reported as 904.5 cGy (standard deviation, ±232.3). Patients treated with lateral fields were more likely to receive lung doses ≥800 cGy (P < .001). The influence of lung dose ≥800 cGy on transplant-related mortality was not significant (hazard ratio [HR], 1.78; P = .21). On univariate analysis, lung dose ≥800 cGy was associated with inferior relapse-free survival (HR, 1.76; P = .04) and OS (HR, 1.85; P = .03). In the multivariate analysis, OS maintained statistical significance (HR, 1.85; P = .04). CONCLUSIONS The variability in TBI techniques resulted in uncertainty with reported lung doses. Lateral fields were associated with higher lung dose, and thus they should be avoided. Patients treated with lung dose <800 cGy in this study had better outcomes. This approach is currently being investigated in the Children's Oncology Group AALL1331 study. Additionally, the Imaging and Radiation Oncology Core Group is evaluating effects of TBI techniques on lung doses using a phantom.
Collapse
Affiliation(s)
| | - Xiaomin Lu
- Children's Oncology Group Data Center, Biostatistics, University of Florida, Gainesville, Florida
| | - Ken Ulin
- Imaging and Radiation Oncology Rhode Island QA Center, Lincoln, Rhode Island
| | - Fran Laurie
- Imaging and Radiation Oncology Rhode Island QA Center, Lincoln, Rhode Island
| | - Sandy Kessel
- Imaging and Radiation Oncology Rhode Island QA Center, Lincoln, Rhode Island
| | - John A Kalapurakal
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | | | - David S Followill
- Imaging and Radiation Oncology Rhode Island QA Center, Houston, Texas
| | | | - Mary K Schmitter
- Imaging and Radiation Oncology Rhode Island QA Center, Lincoln, Rhode Island
| | - Meenakshi Devidas
- Children's Oncology Group Data Center, Biostatistics, University of Florida, Gainesville, Florida
| | - Yichen Chen
- Children's Oncology Group Data Center, Biostatistics, University of Florida, Gainesville, Florida
| | - Donna A Wall
- Manitoba Blood and Marrow Transplant Program, Winnipeg, Manitoba, Canada
| | - Patrick A Brown
- Johns Hopkins University Kimmel Cancer Center, Baltimore, Maryland
| | - Stephen P Hunger
- Children's Hospital of Philadelphia and the Perelman School of Medicine at The University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stephan A Grupp
- Children's Hospital of Philadelphia and the Perelman School of Medicine at The University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael A Pulsipher
- Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California
| |
Collapse
|
34
|
Pan J, Niu Q, Deng B, Liu S, Wu T, Gao Z, Liu Z, Zhang Y, Qu X, Zhang Y, Liu S, Ling Z, Lin Y, Zhao Y, Song Y, Tan X, Zhang Y, Li Z, Yin Z, Chen B, Yu X, Yan J, Zheng Q, Zhou X, Gao J, Chang AH, Feng X, Tong C. CD22 CAR T-cell therapy in refractory or relapsed B acute lymphoblastic leukemia. Leukemia 2019; 33:2854-2866. [PMID: 31110217 DOI: 10.1038/s41375-019-0488-7] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/05/2019] [Accepted: 04/08/2019] [Indexed: 01/08/2023]
Abstract
Despite worldwide promising clinical outcome of CD19 CAR-T therapy, relapse after this therapy is associated with poor prognosis and has become an urgent problem to be solved. We conducted a CD22 CAR T-cell therapy in 34 relapsed or refractory (r/r) B-ALL pediatric and adult patients who failed from previous CD19 CAR T-cell therapy. Complete remission (CR) or CR with incomplete count recovery (CRi) was achieved in 24 of 30 patients (80%) that could be evaluated on day 30 after infusion, which accounted for 70.5% of all 34 enrolled patients. Most patients only experienced mild cytokine-release syndrome and neurotoxicity. Seven CR patients received no further treatment, and 3 of them remained in remission at 6, 6.6, and 14 months after infusion. Eleven CR patients were promptly bridged to transplantation, and 8 of them remained in remission at 4.6 to 13.3 months after transplantation, resulted in 1-year leukemia-free survival rate of 71.6% (95% CI, 44.2-99.0). CD22 antigen loss or mutation was not observed to be associated with relapsed patients. Our study demonstrated that our CD22 CAR T-cells was highly effective in inducing remission in r/r B-ALL patients, and also provided a precious window for subsequent transplantation to achieve durable remission.
Collapse
Affiliation(s)
- Jing Pan
- Department of Hematology, Beijing Boren Hospital, Beijing, 100070, China
| | - Qing Niu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Hematological disorders, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Biping Deng
- Cytology Laboratory, Beijing Boren Hospital, Beijing, 100070, China
| | - Shuangyou Liu
- Department of Hematology, Beijing Boren Hospital, Beijing, 100070, China
| | - Tong Wu
- Department of Bone Marrow Transplantation, Beijing Boren Hospital, Beijing, 100070, China
| | - Zhiyong Gao
- Department of Bone Marrow Transplantation, Beijing Boren Hospital, Beijing, 100070, China
| | - Zhaoli Liu
- Cytology Laboratory, Beijing Boren Hospital, Beijing, 100070, China
| | - Yue Zhang
- Cytology Laboratory, Beijing Boren Hospital, Beijing, 100070, China
| | - Xiaomin Qu
- Cytology Laboratory, Beijing Boren Hospital, Beijing, 100070, China
| | - Yanlei Zhang
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Shaohui Liu
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Zhuojun Ling
- Department of Hematology, Beijing Boren Hospital, Beijing, 100070, China
| | - Yuehui Lin
- Department of Hematology, Beijing Boren Hospital, Beijing, 100070, China
| | - Yongqiang Zhao
- Department of Bone Marrow Transplantation, Beijing Boren Hospital, Beijing, 100070, China
| | - Yanzhi Song
- Department of Bone Marrow Transplantation, Beijing Boren Hospital, Beijing, 100070, China
| | - Xiyou Tan
- Department of Bone Marrow Transplantation, Beijing Boren Hospital, Beijing, 100070, China
| | - Yan Zhang
- Department of Bone Marrow Transplantation, Beijing Boren Hospital, Beijing, 100070, China
| | - Zhihui Li
- Cytology Laboratory, Beijing Boren Hospital, Beijing, 100070, China
| | - Zhichao Yin
- Department of Hematology, Beijing Boren Hospital, Beijing, 100070, China
| | - Bingzhen Chen
- Medical Laboratory, Beijing Boren Hospital, Beijing, 100070, China
| | - Xinjian Yu
- Medical Laboratory, Beijing Boren Hospital, Beijing, 100070, China
| | - Ju Yan
- Medical Laboratory, Beijing Boren Hospital, Beijing, 100070, China
| | - Qinlong Zheng
- Medical Laboratory, Beijing Boren Hospital, Beijing, 100070, China
| | - Xuan Zhou
- Gaobo Healthcare Group, Beijing, China
| | - Jin Gao
- Gaobo Healthcare Group, Beijing, China
| | - Alex H Chang
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| | - Xiaoming Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Hematological disorders, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China. .,Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| | - Chunrong Tong
- Department of Hematology, Beijing Boren Hospital, Beijing, 100070, China.
| |
Collapse
|
35
|
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
|
36
|
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: 2.0] [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
|
37
|
Kinetics and Risk Factors of Relapse after Allogeneic Stem Cell Transplantation in Children with Leukemia: A Long-Term Follow-Up Single-Center Study. Biol Blood Marrow Transplant 2019; 25:100-106. [DOI: 10.1016/j.bbmt.2018.08.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 08/07/2018] [Indexed: 01/13/2023]
|
38
|
Pulsipher MA. Are CAR T cells better than antibody or HCT therapy in B-ALL? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2018; 2018:16-24. [PMID: 30504287 PMCID: PMC6246000 DOI: 10.1182/asheducation-2018.1.16] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Multicenter trials in children and young adults using second-generation CD19-targeted chimeric antigen receptor (CAR) T cells have shown dramatic levels of remission in patients with multiply relapsed/refractory disease (80% to ≥90%). Early results in adult trials have also shown significant responses, and strategies aimed at mitigating toxicities associated with the therapy have improved tolerability. Therefore, if available, CAR T-cell therapy deserves consideration for salvage of children and adults with B-lineage acute lymphoblastic leukemia (B-ALL) who are multiply relapsed, refractory, or relapsed after a previous allogeneic transplantation. For patients with a first relapse or who have persistent minimal residual disease (MRD) after initial or relapse therapy, treatment with blinatumomab or inotuzumab is reasonable to help patients achieve MRD- remission before definitive therapy with allogeneic hematopoietic cell transplantation (HCT). A number of studies in younger patients using 4-1BB-based CAR T-cell constructs lentivirally transduced into patient T cells and then optimally expanded have resulted in long-term persistence without further therapy. In 1 study using CD28-based CARs in adults, the benefit of HCT after CAR T-cell therapy was not clear, because a group of patients experienced long-term remissions without HCT. These data suggest that CAR T-cell therapy may be able to substitute for transplantation in many patients, avoiding the risks and long-term consequences of HCT. With this is mind, and with emerging data better defining ways of enhancing CAR T-cell persistence and avoiding relapse through antigen escape, CAR T cells will have a growing role in treatment of both pediatric and adult B-ALLs in the coming years.
Collapse
Affiliation(s)
- Michael A Pulsipher
- University of Southern California Keck School of Medicine, Children's Hospital Los Angeles, Los Angeles CA
| |
Collapse
|
39
|
CAR-T Cell Therapy for Acute Lymphoblastic Leukemia: Transforming the Treatment of Relapsed and Refractory Disease. Curr Hematol Malig Rep 2018; 13:396-406. [DOI: 10.1007/s11899-018-0470-x] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
40
|
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: 0.5] [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
|
41
|
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: 158] [Impact Index Per Article: 26.3] [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
|
42
|
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: 43] [Impact Index Per Article: 7.2] [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
|
43
|
Page KM, Labopin M, Ruggeri A, Michel G, Diaz de Heredia C, O'Brien T, Picardi A, Ayas M, Bittencourt H, Vora AJ, Troy J, Bonfim C, Volt F, Gluckman E, Bader P, Kurtzberg J, Rocha V. Factors Associated with Long-Term Risk of Relapse after Unrelated Cord Blood Transplantation in Children with Acute Lymphoblastic Leukemia in Remission. Biol Blood Marrow Transplant 2017; 23:1350-1358. [PMID: 28438676 PMCID: PMC5569913 DOI: 10.1016/j.bbmt.2017.04.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 04/16/2017] [Indexed: 12/20/2022]
Abstract
For pediatric patients with acute lymphoblastic leukemia (ALL), relapse is an important cause of treatment failure after unrelated cord blood transplant (UCBT). Compared with other donor sources, relapse is similar or even reduced after UCBT despite less graft-versus-host disease (GVHD). We performed a retrospective analysis to identify risk factors associated with the 5-year cumulative incidence of relapse after UCBT. In this retrospective, registry-based study, we examined the outcomes of 640 children (<18 years) with ALL in first complete remission (CR1; n = 257, 40%) or second complete remission (CR2; n = 383, 60%) who received myeloablative conditioning followed by a single-unit UCBT from 2000 to 2012. Most received antithymocyte globulin (88%) or total body irradiation (TBI; 69%), and cord blood grafts were primarily mismatched at 1 (50%) or 2+ (34%) HLA loci. Considering patients in CR1, the rates of 5-year overall survival (OS), leukemia-free survival (LFS), and relapse were 59%, 52%, and 23%, respectively. In multivariate analysis (MVA), acute GVHD (grades II to IV) and TBI protected against relapse. In patients in CR2, rates of 5-year OS, LFS, and the cumulative incidence of relapse were 46%, 44%, and 28%, respectively. In MVA, longer duration from diagnosis to UCBT (≥30 months) and TBI were associated with decreased relapse risk. Importantly, receiving a fully HLA matched graft was a strong risk factor for increased relapse in MVA. An exploratory analysis of all 640 patients supported the important association between the presence of acute GVHD and less relapse but also demonstrated an increased risk of nonrelapse mortality. In conclusion, the impact of GVHD as a graft-versus-leukemia marker is evident in pediatric ALL after UCBT. Strategies that promote graft-versus-leukemia while harnessing GVHD should be further investigated.
Collapse
Affiliation(s)
- Kristin M Page
- Division of Pediatric Blood and Marrow Transplantation, Duke University Medical Center, Durham, North Carolina.
| | - Myriam Labopin
- EBMT, Acute Leukemia Working Party, Service d'hematologie et therapie cellulaire, Hôpital Saint Antoine, Paris, France
| | - Annalisa Ruggeri
- EBMT, Acute Leukemia Working Party, Service d'hematologie et therapie cellulaire, Hôpital Saint Antoine, Paris, France; Eurocord, Hospital Saint Louis APHP, University Paris-Diderot, Paris, France; Monacord, Centre Scientifique de Monaco, Monaco-Ville, Monaco
| | - Gerard Michel
- Timone Enfants Hospital and Aix-Marseille University, Department of Pediatric Hematology and Oncology, Marseille, France
| | | | - Tracey O'Brien
- Blood and Marrow Transplant Program, Sydney Children's Hospital, Randwick, New South Wales, Australia
| | | | - Mouhab Ayas
- Department of Pediatric Hematology/Oncology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | | | - Ajay J Vora
- Department of Pediatric Haematology, The Children's Hospital, Sheffield, UK; Department of Haematology and Oncology, Great Ormond Street Hospital, London, UK
| | - Jesse Troy
- Division of Pediatric Blood and Marrow Transplantation, Duke University Medical Center, Durham, North Carolina
| | - Carmen Bonfim
- Hospital Das Clinicas, Universidade Federal do Parana, Curitiba, Brazil
| | - Fernanda Volt
- Eurocord, Hospital Saint Louis APHP, University Paris-Diderot, Paris, France; Monacord, Centre Scientifique de Monaco, Monaco-Ville, Monaco
| | - Eliane Gluckman
- Eurocord, Hospital Saint Louis APHP, University Paris-Diderot, Paris, France; Monacord, Centre Scientifique de Monaco, Monaco-Ville, Monaco
| | - Peter Bader
- Division for Stem Cell Transplantation and Immunology, Hospital for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Joanne Kurtzberg
- Division of Pediatric Blood and Marrow Transplantation, Duke University Medical Center, Durham, North Carolina
| | - Vanderson Rocha
- Eurocord, Hospital Saint Louis APHP, University Paris-Diderot, Paris, France; Monacord, Centre Scientifique de Monaco, Monaco-Ville, Monaco; Hospital Das Clinicas, University of Sao Paulo, Sao Paulo, Brazil; Churchill Hospital, Oxford University, Oxford, UK
| |
Collapse
|
44
|
Lamble A, Phelan R, Burke M. When Less Is Good, Is None Better? The Prognostic and Therapeutic Significance of Peri-Transplant Minimal Residual Disease Assessment in Pediatric Acute Lymphoblastic Leukemia. J Clin Med 2017; 6:E66. [PMID: 28686179 PMCID: PMC5532574 DOI: 10.3390/jcm6070066] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/26/2017] [Accepted: 07/03/2017] [Indexed: 02/06/2023] Open
Abstract
The measurement of minimal residual disease (MRD) in pediatric acute lymphoblastic leukemia (ALL) has become the most important prognostic tool of, and the backbone to, upfront risk stratification. While MRD assessment is the standard of care for assessing response and predicting outcomes for pediatric patients with ALL receiving chemotherapy, its use in allogeneic hematopoietic stem cell transplant (HSCT) has been less clearly defined. Herein, we discuss the importance of MRD assessment during the peri-HSCT period and its role in prognostication and management.
Collapse
Affiliation(s)
- Adam Lamble
- Pediatric Hematology/Oncology, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Rachel Phelan
- Pediatric Hematology/Oncology/Blood and Marrow Transplant, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Michael Burke
- Pediatric Hematology/Oncology/Blood and Marrow Transplant, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
45
|
Pierro J, Hogan LE, Bhatla T, Carroll WL. New targeted therapies for relapsed pediatric acute lymphoblastic leukemia. Expert Rev Anticancer Ther 2017. [PMID: 28649891 DOI: 10.1080/14737140.2017.1347507] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION The improvement in outcomes for children with acute lymphoblastic leukemia (ALL) is one of the greatest success stories of modern oncology however the prognosis for patients who relapse remains dismal. Recent discoveries by high resolution genomic technologies have characterized the biology of relapsed leukemia, most notably pathways leading to the drug resistant phenotype. These observations open the possibility of targeting such pathways to prevent and/or treat relapse. Likewise, early experiences with new immunotherapeutic approaches have shown great promise. Areas covered: We performed a literature search on PubMed and recent meeting abstracts using the keywords below. We focused on the biology and clonal evolution of relapsed disease highlighting potential new targets of therapy. We further summarized the results of early trials of the three most prominent immunotherapy agents currently under investigation. Expert commentary: Discovery of targetable pathways that lead to drug resistance and recent breakthroughs in immunotherapy show great promise towards treating this aggressive disease. The best way to treat relapse, however, is to prevent it which makes incorporation of these new approaches into frontline therapy the best approach. Challenges remain to balance efficacy with toxicity and to prevent the emergence of resistant subclones which is why combining these newer agents with conventional chemotherapy will likely become standard of care.
Collapse
Affiliation(s)
- Joanna Pierro
- a Division of Pediatric Hematology Oncology, Department of Pediatrics , Perlmutter Cancer Center, NYU Langone Medical Center , New York , NY , USA
| | - Laura E Hogan
- b Division of Pediatric Hematology/Oncology, Department of Pediatrics , Stony Brook Children's , Stony Brook , NY , USA
| | - Teena Bhatla
- a Division of Pediatric Hematology Oncology, Department of Pediatrics , Perlmutter Cancer Center, NYU Langone Medical Center , New York , NY , USA
| | - William L Carroll
- a Division of Pediatric Hematology Oncology, Department of Pediatrics , Perlmutter Cancer Center, NYU Langone Medical Center , New York , NY , USA
| |
Collapse
|
46
|
Minimal residual disease assessment by next-generation sequencing. Better tools to gaze into the crystal ball? Bone Marrow Transplant 2017; 52:952-954. [PMID: 28581473 DOI: 10.1038/bmt.2017.104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 04/24/2017] [Accepted: 04/26/2017] [Indexed: 11/08/2022]
|
47
|
Umeda K, Iwai A, Kawaguchi K, Mikami M, Nodomi S, Saida S, Hiramatsu H, Heike T, Ohmori K, Adachi S. Impact of post-transplant minimal residual disease on the clinical outcome of pediatric acute leukemia. Pediatr Transplant 2017; 21. [PMID: 28370903 DOI: 10.1111/petr.12926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/24/2017] [Indexed: 11/27/2022]
Abstract
This retrospective study examined the clinical significance of FCM-MRD in 36 patients with ALL and 29 patients with AML after their first allogeneic HSCT. Hematological (FCM-MRD ≥5.0%) and molecular relapse (FCM-MRD <5.0%) were first detected in 10 and two patients with ALL and in seven and eight patients with AML, respectively. Eight of 10 patients with molecular relapse eventually progressed to hematological relapse, although most were treated with immunological intervention by aggressive discontinuation of immunosuppressive therapy or donor lymphocyte infusion. Among these 12 patients, four of seven patients that obtained MRDneg CR following post-transplant chemotherapy remain alive and disease-free after their second HSCT; however, all five patients who underwent a second HSCT in non-CR died of disease or treatment-related complications. As the FCM-MRD monitoring system used in the current study was probably not sensitive enough to detect MRD, which could be elucidated by immunological intervention, more sensitive diagnostic tools are mandatory for post-transplant MRD monitoring. Additional studies are required to address the impact of presecond transplant MRD on the clinical outcome of second HSCT.
Collapse
Affiliation(s)
- Katsutsugu Umeda
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Atsushi Iwai
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koji Kawaguchi
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masamitsu Mikami
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Seishiro Nodomi
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Saida
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hidefumi Hiramatsu
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshio Heike
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Katsuyuki Ohmori
- Department of Clinical Laboratory, Kyoto University Hospital, Kyoto, Japan
| | - Souichi Adachi
- Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
48
|
Pan J, Yang JF, Deng BP, Zhao XJ, Zhang X, Lin YH, Wu YN, Deng ZL, Zhang YL, Liu SH, Wu T, Lu PH, Lu DP, Chang AH, Tong CR. High efficacy and safety of low-dose CD19-directed CAR-T cell therapy in 51 refractory or relapsed B acute lymphoblastic leukemia patients. Leukemia 2017; 31:2587-2593. [DOI: 10.1038/leu.2017.145] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 04/25/2017] [Accepted: 05/04/2017] [Indexed: 12/18/2022]
|
49
|
Mo XD, Zhang XH, Xu LP, Wang Y, Yan CH, Chen H, Chen YH, Han W, Wang FR, Wang JZ, Liu KY, Huang XJ. Comparison of outcomes after donor lymphocyte infusion with or without prior chemotherapy for minimal residual disease in acute leukemia/myelodysplastic syndrome after allogeneic hematopoietic stem cell transplantation. Ann Hematol 2017; 96:829-838. [PMID: 28285386 DOI: 10.1007/s00277-017-2960-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 02/16/2017] [Indexed: 11/24/2022]
Abstract
The efficacy of donor lymphocyte infusion (DLI) without chemotherapy was investigated and compared with that of chemotherapy prior to DLI (Chemo-DLI) in patients who were minimal residual disease (MRD)-positive after allogeneic hematopoietic stem cell transplantation (HSCT). We enrolled 115 consecutive patients who received either DLI (n = 20) or Chemo-DLI (n = 95) during the same period. For each DLI recipient, three recipients matched for age at the HSCT, underlying diseases, and the year of the HSCT were randomly selected from the Chemo-DLI cohort (n = 60). The 2-year cumulative incidence of severe acute graft-versus-host disease (GVHD) and chronic GVHD was comparable between the groups. Fifteen (75.0%) and 47 (78.3%) patients in the DLI and Chemo-DLI groups turned MRD-negative, respectively. The 2-year cumulative incidences of relapse and non-relapse mortality after intervention were 30.7 versus 39.6% (P = 0.582) and 10.3 versus 6.0% (P = 0.508) in the DLI and Chemo-DLI groups, respectively. The 2-year probabilities of disease-free, overall, and GVHD-free/relapse-free survival after preemptive intervention were 58.9 versus 54.3% (P = 0.862), 69.3 versus 78.1% (P = 0.361), and 44.4 versus 35.1% (P = 0.489) in the DLI and Chemo-DLI groups, respectively. In multivariate analysis, the intervention method did not significantly influence the clinical outcomes. In summary, preemptive DLI alone may be effective for patients who are MRD-positive and may be a potential alternative for patients who refuse or are unable to receive Chemo-DLI after HSCT.
Collapse
Affiliation(s)
- Xiao-Dong Mo
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China
| | - Lan-Ping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China
| | - Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China
| | - Chen-Hua Yan
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China
| | - Huan Chen
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China
| | - Yu-Hong Chen
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China
| | - Wei Han
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China
| | - Feng-Rong Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China
| | - Jing-Zhi Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China
| | - Kai-Yan Liu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China. .,Peking-Tsinghua Center for Life Sciences, Beijing, China. .,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.
| |
Collapse
|
50
|
Liu J, Zhong JF, Zhang X, Zhang C. Allogeneic CD19-CAR-T cell infusion after allogeneic hematopoietic stem cell transplantation in B cell malignancies. J Hematol Oncol 2017; 10:35. [PMID: 28143567 PMCID: PMC5282795 DOI: 10.1186/s13045-017-0405-3] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/19/2017] [Indexed: 12/22/2022] Open
Abstract
Background Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is considered the cornerstone in treatment of hematological malignancies. However, relapse of the hematological disease after allo-HSCT remains a challenge and is associated with poor long-term survival. Chimeric antigen receptor redirected T cells (CAR-T cells) can lead to disease remission in patients with relapsed/refractory hematological malignancies. However, the therapeutic window for infusion of CAR-T cells post allo-HSCT and its efficacy are debatable. Main body In this review, we first discuss the use of CAR-T cells for relapsed cases after allo-HSCT. We then review the toxicities and the occurrence of graft-versus-host disease in relapsed patients who received CAR-T cells post allo-HSCT. Finally, we review clinical trial registrations and the therapeutic time window for infusion of CAR-T cells post allo-HSCT. Conclusions The treatment of allogeneic CAR-T cells is beneficial for patients with relapsed B cell malignancies after allo-HSCT with low toxicities and complications. However, multicenter clinical trials with larger sample sizes should be performed to select the optimal therapeutic window and confirm its efficacy.
Collapse
Affiliation(s)
- Jun Liu
- Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China
| | - Jiang F Zhong
- Division of Periodontology, Diagnostic Sciences & Dental Hygiene, and Division of Biomedical Sciences, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Xi Zhang
- Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China
| | - Cheng Zhang
- Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China.
| |
Collapse
|