1
|
Shah NN, Schafer ES, Chi YY, Malvar J, Heym KM, Place AE, Burns M, Chang BH, Slone T, Verma A, Gossai N, Shaw PH, Burke MJ, Hermiston M, Schore RJ, Cooper T, Pauly M, Rushing T, Jarosinski P, Florendo E, Yates B, Widemann BC, Peer CJ, Figg WD, Silverman LB, Bhojwani D, Wayne AS. Vincristine Sulfate Liposome Injection With Combination Chemotherapy for Children, Adolescents, and Young Adults With Relapsed Acute Lymphoblastic Leukemia: A Therapeutic Advances in Childhood Leukemia and Lymphoma Consortium Trial. Pediatr Blood Cancer 2025; 72:e31584. [PMID: 39937083 DOI: 10.1002/pbc.31584] [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: 12/09/2024] [Revised: 01/12/2025] [Accepted: 01/18/2025] [Indexed: 02/13/2025]
Abstract
INTRODUCTION Vincristine sulfate liposome injection (VSLI), a liposomal formulation of vincristine, may be better tolerated than standard aqueous vincristine and enable dose intensification. PROCEDURES Based on single-agent tolerability, activity, and FDA approval in adults with acute lymphoblastic leukemia (ALL), we tested the safety and feasibility of VSLI as replacement for standard vincristine in the UK ALL R3 mitoxantrone-based four-drug induction (Cohort A), a three-drug anthracycline-free induction (Cohort B), and maintenance chemotherapy (Cohort C) in children and young adults with relapsed/refractory B-cell ALL. RESULTS Among 29 participants with a median age of 12.4 years (range: 1.8-19.6 years), 16 received Cohort A, eight received Cohort B, and five received Cohort C therapy. Dose level 1 (DL1): 1.5 mg/m2 and dose level 2 (DL2): 2 mg/m2 of VSLI, each without a dose cap, were tested. Collectively, the median VSLI dose administered was 1.9 mg (range: 0.71-4.06 mg), and 13 (44.8%) received a dose above the standard 2 mg vincristine dose cap. Dose-limiting toxicities (DLTs) at DL2 were seen in three patients, two in Cohort A and one in Cohort B, prompting further evaluation at DL1 for both cohorts. No DLTs were experienced at DL1. Only DL2 was tested in Cohort C-without DLT. Complete remissions were seen in 14 of 16 (87.5%) participants in Cohort A; three of eight (37.5%) in Cohort B; and one (20%) in Cohort C. VSLI with combination chemotherapy at DL1 was generally well tolerated. CONCLUSION Based on the promising response signal in this heavily pretreated population, further study of VSLI is warranted. (ClinicalTrials.gov NCT02879643).
Collapse
Affiliation(s)
- Nirali N Shah
- Pediatric Oncology Branch, National Cancer Institute/Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Eric S Schafer
- Division of Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Cancer and Hematology Center, Houston, Texas, USA
| | - Yueh-Yun Chi
- Division of Hematology-Oncology, Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jemily Malvar
- Division of Hematology-Oncology, Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Kenneth M Heym
- Hematology and Oncology, Cook Children's Medical Center, Fort Worth, Texas, USA
| | - Andrew E Place
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Melissa Burns
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Bill H Chang
- Division of Hematology and Oncology, Department of Pediatrics, Oregon Health & Science University, Portland, Oregon, USA
| | - Tamra Slone
- Children's Medical Center, UT Southwestern, Dallas, Texas, USA
| | - Anupam Verma
- Pediatric Specialists of Virginia, Fairfax, Virginia, USA
- Division of Pediatric Hematology Oncology, Primary Children's Hospital, University of Utah, Salt Lake City, Utah, USA
| | - Nathan Gossai
- Center for Cancer and Blood Disorders, Children's Minnesota, Minneapolis, Minnesota, USA
| | - Peter H Shaw
- Division of Hematology and Oncology, Department of Pediatrics, Children's Wisconsin, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Michael J Burke
- Division of Hematology and Oncology, Department of Pediatrics, Children's Wisconsin, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Michelle Hermiston
- Pediatric Hematology/Oncology, Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Reuven J Schore
- Division of Pediatric Oncology, Children's National Hospital/George Washington University SMHS, Washington, District of Columbia, USA
| | - Todd Cooper
- Seattle Children's Cancer and Blood Disorders Center, University of Washington, Seattle, Washington, USA
| | - Melinda Pauly
- Department of Pediatric Hematology Oncology, Emory University and Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Teresa Rushing
- Division of Hematology-Oncology, Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Department of Pharmacy, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Paul Jarosinski
- Pediatric Oncology Branch, National Cancer Institute/Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Ellynore Florendo
- Division of Hematology-Oncology, Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Bonnie Yates
- Pediatric Oncology Branch, National Cancer Institute/Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Brigitte C Widemann
- Pediatric Oncology Branch, National Cancer Institute/Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Cody J Peer
- Clinical Pharmacology Laboratory, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, Maryland, USA
| | - William D Figg
- Clinical Pharmacology Laboratory, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Lewis B Silverman
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Columbia University Medical Center, New York, New York, USA
| | - Deepa Bhojwani
- Division of Hematology-Oncology, Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Alan S Wayne
- Division of Hematology-Oncology, Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
2
|
Atre T, Nguyen V, Chow V, Reid GSD, Vercauteren S. A Comparative Study of B Cell Blast Isolation Methods from Bone Marrow Aspirates of Pediatric Leukemia Patients. Biopreserv Biobank 2025; 23:46-52. [PMID: 38686645 DOI: 10.1089/bio.2023.0133] [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] [Indexed: 05/02/2024] Open
Abstract
Density gradient centrifugation is a conventional technique widely utilized to isolate bone marrow mononuclear cells (BM-MNC) from bone marrow (BM) aspirates obtained from pediatric B-cell acute lymphoblastic leukemia (B-ALL) patients. Nevertheless, this technique achieves incomplete recovery of mononuclear cells and is relatively time-consuming and expensive. Given that B-ALL is the most common childhood malignancy, alternative methods for processing B-ALL samples may be more cost-effective. In this pilot study, we use several readouts, including immune phenotype, cell viability, and leukemia-initiating capacity in immune-deficient mice, to directly compare the density gradient centrifugation and buffy coat processing methods. Our findings indicate that buffy coat isolation yields comparable BM-MNC product in terms of both immune and leukemia cell content and could provide a viable, lower cost alternative for biobanks processing pediatric leukemia samples.
Collapse
Affiliation(s)
- Tanmaya Atre
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, Canada
| | - Vi Nguyen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
- BC Children's Hospital BioBank, BC Children's Hospital, Vancouver, Canada
| | - Veronica Chow
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
- BC Children's Hospital BioBank, BC Children's Hospital, Vancouver, Canada
| | - Gregor S D Reid
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Suzanne Vercauteren
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
- BC Children's Hospital BioBank, BC Children's Hospital, Vancouver, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
- Division of Hematopathology, BC Children's Hospital, Vancouver, Canada
| |
Collapse
|
3
|
Bhatla T, Cooper S, Hogan LE. Low-risk relapsed acute lymphoblastic leukemia in children and young adults: what have we learnt and what's next? Leuk Lymphoma 2024; 65:1398-1404. [PMID: 38861360 DOI: 10.1080/10428194.2024.2362408] [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: 04/04/2024] [Accepted: 05/27/2024] [Indexed: 06/13/2024]
Abstract
While outcomes for newly diagnosed children with acute lymphoblastic leukemia (ALL) have improved over the last few decades, 10-15% will relapse. Outcomes for those children with relapse remains a challenge, with 5-year overall survival of approximately 35-60%. Large cooperative group trials have identified factors associated with favorable (low risk, LR) outcome at relapse, including later relapse, B-cell phenotype, isolated extramedullary relapse and a good response to initial re-induction therapy. Contemporary therapeutic regimens are aimed at improving outcomes, while decreasing toxicity. A main focus of current research involves how immunotherapy can be best incorporated with cytotoxic chemotherapy to improve survival in relapsed ALL. Here we review therapeutic strategies for LR relapse, including review of recently completed and ongoing trials.
Collapse
Affiliation(s)
- Teena Bhatla
- Children's Hospital of New Jersey at Newark Beth Israel, Newark, NJ, USA
| | - Stacy Cooper
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - Laura E Hogan
- Department of Pediatrics, Stony Brook Children's, Stony Brook, NY, USA
| |
Collapse
|
4
|
Uittenboogaard A, van de Velde M, van de Heijden L, Mukuhi L, de Vries N, Langat S, Olbara G, Huitema ADR, Vik T, Kaspers G, Njuguna F. Vincristine exposure in Kenyan children with cancer: CHAPATI feasibility study. Pediatr Blood Cancer 2024; 71:e31160. [PMID: 38956809 DOI: 10.1002/pbc.31160] [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: 02/08/2024] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 07/04/2024]
Abstract
The low incidence of vincristine-induced peripheral neuropathy (VIPN) in Kenyan children may result from low vincristine exposure. We studied vincristine exposure in Kenyan children and dose-escalated in case of low vincristine exposure (NCT05844670). Average vincristine exposure was high. Individual vincristine exposure was assessed with a previously developed nomogram. A 20% dose increase was recommended for participants with low exposure and no VIPN, hyperbilirubinemia, or malnutrition. None of the 15 participants developed VIPN. Low vincristine exposure was seen in one participant: a dose increase was implemented without side effects. In conclusion, the participants did not develop VIPN despite having high vincristine exposure.
Collapse
Affiliation(s)
- Aniek Uittenboogaard
- Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Mirjam van de Velde
- Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Lisa van de Heijden
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Clinical Pharmacy, OLVG, Amsterdam, The Netherlands
| | - Leah Mukuhi
- Academic Model Providing Access to Healthcare (AMPATH), Eldoret, Kenya
| | - Niels de Vries
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sandra Langat
- Academic Model Providing Access to Healthcare (AMPATH), Eldoret, Kenya
| | - Gilbert Olbara
- Department of Child Health and Paediatrics, Moi University/Moi Teaching and Referral Hospital, Eldoret, Kenya
| | - Alwin D R Huitema
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Terry Vik
- Pediatric Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Gertjan Kaspers
- Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Festus Njuguna
- Department of Child Health and Paediatrics, Moi University/Moi Teaching and Referral Hospital, Eldoret, Kenya
| |
Collapse
|
5
|
Ueki H, Ogawa C, Goto H, Nishi M, Yamanaka J, Mochizuki S, Nishikawa T, Kumamoto T, Nishiuchi R, Kikuta A, Yamamoto S, Igarashi S, Sato A, Hori T, Saito AM, Watanabe T, Deguchi T, Manabe A, Horibe K, Toyoda H. TBI, etoposide, and cyclophosphamide conditioning for intermediate-risk relapsed childhood acute lymphoblastic leukemia. Int J Hematol 2024; 119:450-458. [PMID: 38267673 DOI: 10.1007/s12185-024-03710-6] [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: 04/26/2023] [Revised: 12/27/2023] [Accepted: 01/08/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND In children with intermediate-risk relapsed acute lymphoblastic leukemia (ALL), allogeneic hematopoietic stem cell transplantation (allo-HSCT) has markedly improved the outcome of patients with an unsatisfactory minimal residual disease (MRD) response. Total body irradiation (TBI), etoposide (ETP), and cyclophosphamide (CY) have been shown to be equivalent to or better than TBI + ETP for conditioning, so we hypothesized that even greater survival could be achieved due to recent advances in HSCT and supportive care. PROCEDURE We prospectively analyzed the efficacy and safety of allo-HSCT with a unified conditioning regimen of TBI + ETP + CY in children with intermediate-risk relapsed ALL, based on MRD in the bone marrow after induction, from the Japanese Pediatric Leukemia/Lymphoma Study Group (JPLSG) ALL-R08-II nationwide cohort (UMIN000002025). RESULTS Twenty patients with post-induction MRD ≥ 10-3 and two not evaluated for MRD underwent allo-HSCT. Engraftment was confirmed in all patients, and no transplantation-related mortality was observed. The 3-year event-free survival and overall survival rates after transplantation were 86.4% ± 7.3% and 95.5% ± 4.4%, respectively. CONCLUSION Allo-HSCT based on post-induction MRD with TBI + ETP + CY conditioning was feasible in Japanese children with intermediate-risk relapsed ALL.
Collapse
Affiliation(s)
- Hideaki Ueki
- Department of Pediatric Hematology/Oncology, Japanese Red Cross Narita Hospital, Narita, Japan
| | - Chitose Ogawa
- Department of Pediatric Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroaki Goto
- Division of Hematology/Oncology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Masanori Nishi
- Department of Pediatrics, Faculty of Medicine, Saga University, Saga, Japan
| | - Junko Yamanaka
- Department of Pediatrics, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shinji Mochizuki
- Department of Pediatrics, National Center for Global Health and Medicine, Tokyo, Japan
| | - Takuro Nishikawa
- Department of Pediatrics, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Tadashi Kumamoto
- Department of Pediatric Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Ritsuo Nishiuchi
- Department of Pediatrics, Kochi Health Sciences Center, Kochi, Japan
| | - Atsushi Kikuta
- Department of Pediatric Oncology, Fukushima Medical University Hospital, Fukushima, Japan
| | - Shohei Yamamoto
- Department of Pediatrics, Tokai University School of Medicine, Isehara, Japan
| | - Shunji Igarashi
- Department of Pediatric Hematology/Oncology, Japanese Red Cross Narita Hospital, Narita, Japan
| | - Atsushi Sato
- Department of Hematology/Oncology, Miyagi Children's Hospital, Sendai, Japan
| | - Toshinori Hori
- Department of Pediatrics, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Akiko M Saito
- Clinical Research Center, NHO Nagoya Medical Center, Nagoya, Japan
| | - Tomoyuki Watanabe
- Department of Nutritional Science, Faculty of Psychological and Physical Science, Aichi Gakuin University, Nisshin, Japan
| | - Takao Deguchi
- Division of Cancer Immunodiagnostics, Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Atsushi Manabe
- Department of Pediatrics, Hokkaido University, Sapporo, Japan
| | - Keizo Horibe
- Clinical Research Center, NHO Nagoya Medical Center, Nagoya, Japan
| | - Hidemi Toyoda
- Department of Pediatrics, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan.
| |
Collapse
|
6
|
Murphy L, Aldoss I. Blinatumomab improves outcomes for pediatric patients with low-risk B-cell acute lymphoblastic leukemia in first marrow relapse. Transl Pediatr 2024; 13:530-534. [PMID: 38590377 PMCID: PMC10998983 DOI: 10.21037/tp-23-521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 12/29/2023] [Indexed: 04/10/2024] Open
Affiliation(s)
- Lindsey Murphy
- Department of Pediatrics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Ibrahim Aldoss
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
7
|
Sidhu J, Gogoi MP, Krishnan S, Saha V. Relapsed Acute Lymphoblastic Leukemia. Indian J Pediatr 2024; 91:158-167. [PMID: 37341952 DOI: 10.1007/s12098-023-04635-4] [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: 02/09/2023] [Accepted: 04/21/2023] [Indexed: 06/22/2023]
Abstract
Outcomes for children with acute lymphoblastic leukemia (ALL) have improved worldwide to >85%. For those who relapse, outcomes have remained static at ~50% making relapsed acute lymphoblastic leukemia one of the leading causes of death in childhood cancers. Those relapsing within 18 mo in the bone marrow have a particularly dismal outcome. The mainstay of treatment is chemotherapy, local radiotherapy with or without hematopoietic stem cell transplantation (HSCT). Improved biological understanding of mechanisms of relapse and drug resistance, use of innovative strategies to identify the most effective and least toxic treatment regimens and global partnerships are needed to improve outcomes in these patients. Over the last decade, new therapeutic options and strategies have been developed for relapsed ALL including immunotherapies and cellular therapies. It is imperative to understand how and when to use these newer approaches in relapsed ALL. Increasingly, integrated precision oncology strategies are being used to individualize treatment of patients with relapsed ALL, especially in patients with poor response disease.
Collapse
Affiliation(s)
- Jasmeet Sidhu
- Department of Pediatric Hematology and Oncology, Tata Medical Center, Kolkata, 700160, India
- Tata Translational Cancer Research Center, Tata Medical Center, Kolkata, 700160, India
- University Children's Hospital, Zurich, 8008, Switzerland
| | - Manash Pratim Gogoi
- Tata Translational Cancer Research Center, Tata Medical Center, Kolkata, 700160, India
| | - Shekhar Krishnan
- Department of Pediatric Hematology and Oncology, Tata Medical Center, Kolkata, 700160, India
- Tata Translational Cancer Research Center, Tata Medical Center, Kolkata, 700160, India
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M20 4BX, UK
| | - Vaskar Saha
- Department of Pediatric Hematology and Oncology, Tata Medical Center, Kolkata, 700160, India.
- Tata Translational Cancer Research Center, Tata Medical Center, Kolkata, 700160, India.
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M20 4BX, UK.
| |
Collapse
|
8
|
Barsan V, Li Y, Prabhu S, Baggott C, Nguyen K, Pacenta H, Phillips CL, Rossoff J, Stefanski H, Talano JA, Moskop A, Baumeister S, Verneris MR, Myers GD, Karras NA, Cooper S, Qayed M, Hermiston M, Satwani P, Krupski C, Keating A, Fabrizio V, Chinnabhandar V, Kunicki M, Curran KJ, Mackall CL, Laetsch TW, Schultz LM. Tisagenlecleucel utilisation and outcomes across refractory, first relapse and multiply relapsed B-cell acute lymphoblastic leukemia: a retrospective analysis of real-world patterns. EClinicalMedicine 2023; 65:102268. [PMID: 37954907 PMCID: PMC10632672 DOI: 10.1016/j.eclinm.2023.102268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 09/19/2023] [Accepted: 09/26/2023] [Indexed: 11/14/2023] Open
Abstract
Background Tisagenlecleucel was approved by the Food and Drug Administration (FDA) in 2017 for refractory B-cell acute lymphoblastic leukemia (B-ALL) and B-ALL in ≥2nd relapse. Outcomes of patients receiving commercial tisagenlecleucel upon 1st relapse have yet to be established. We aimed to report real-world tisagenlecleucel utilisation patterns and outcomes across indications, specifically including patients treated in 1st relapse, an indication omitted from formal FDA approval. Methods We conducted a retrospective analysis of real-world tisagenlecleucel utilisation patterns across 185 children and young adults treated between August 30, 2017 and March 6, 2020 from centres participating in the Pediatric Real-World CAR Consortium (PRWCC), within the United States. We described definitions of refractory B-ALL used in the real-world setting and categorised patients by reported Chimeric Antigen Receptor (CAR) T-cell indication, including refractory, 1st relapse and ≥2nd relapse B-ALL. We analysed baseline patient characteristics and post-tisagenlecleucel outcomes across defined cohorts. Findings Thirty-six percent (n = 67) of our cohort received tisagenlecleucel following 1st relapse. Of 66 evaluable patients, 56 (85%, 95% CI 74-92%) achieved morphologic complete response. Overall-survival (OS) and event-free survival (EFS) at 1-year were 69%, (95% CI 58-82%) and 49%, (95% CI 37-64%), respectively, with survival outcomes statistically comparable to remaining patients (OS; p = 0.14, EFS; p = 0.39). Notably, toxicity was increased in this cohort, warranting further study. Interestingly, of 30 patients treated for upfront refractory disease, 23 (77%, 95% CI 58-90%) had flow cytometry and/or next-generation sequencing (NGS) minimum residual disease (MRD)-only disease at the end of induction, not meeting the historic morphologic definition of refractory. Interpretation Our findings suggested that tisagenlecleucel response and survival rates overlap across patients treated with upfront refractory B-ALL, B-ALL ≥2nd relapse and B-ALL in 1st relapse. We additionally highlighted that definitions of refractory B-ALL are evolving beyond morphologic measures of residual disease. Funding St. Baldrick's/Stand Up 2 Cancer, Parker Institute for Cancer Immunotherapy, Virginia and D.K. Ludwig Fund for Cancer Research.
Collapse
Affiliation(s)
- Valentin Barsan
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, 1000 Welch Road, Suite 300, Palo Alto, CA 94304, USA
| | - Yimei Li
- Department of Pediatrics, Children's Hospital of Philadelphia/University of Pennsylvania, 3401 Civic Center Blvd., Philadelphia, PA 19104, USA
- Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA 19104, USA
| | - Snehit Prabhu
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, 1000 Welch Road, Suite 300, Palo Alto, CA 94304, USA
| | - Christina Baggott
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, 1000 Welch Road, Suite 300, Palo Alto, CA 94304, USA
| | - Khanh Nguyen
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, 1000 Welch Road, Suite 300, Palo Alto, CA 94304, USA
| | - Holly Pacenta
- Cook Children’s Hospital, 1500 Cooper St 5th Floor, Fort Worth, TX 76104, USA
- Department of Pediatrics, The University of Texas Southwestern Medical Center/Children’s Health, 5323 Harry Hines Blvd., Dallas, TX 75390-9063, USA
| | - Christine L. Phillips
- Department of Pediatrics, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH 45229-3026, USA
- Cincinnati Children’s Hospital Medical Center, Cancer and Blood Disease Institute, 3333 Burnet Avenue, Cincinnati, OH 45229-3026, USA
| | - Jenna Rossoff
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago, 225 E Chicago Ave, Chicago, IL 60611, USA
| | - Heather Stefanski
- Division of Pediatric Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota Medical School, 2450 Riverside Ave S AO-102, Minneapolis, MN 55454, USA
| | - Julie-An Talano
- Department of Pediatric Hematology Oncology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Amy Moskop
- Department of Pediatric Hematology Oncology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Susanne Baumeister
- Dana Farber/Boston Children’s Hospital, 450 Brookline Avenue Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA 02115, USA
| | - Michael R. Verneris
- University of Colorado, Anschutz Medical Campus, Colorado Children’s Hospital, 13123 East 16th Avenue, Aurora, CO 80045, USA
| | | | - Nicole A. Karras
- Department of Pediatrics, City of Hope National Medical Center, 1500 E Duarte Rd, Duarte, CA 91010, USA
| | - Stacy Cooper
- Department of Oncology, Sidney Kimmel Cancer Center at John Hopkins School of Medicine, Baltimore, MD, USA
| | - Muna Qayed
- Emory University and Children’s Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA 30322, USA
| | - Michelle Hermiston
- University of California San Francisco Benioff Children’s Hospital, 1975 4th St., San Francisco, CA 94158, USA
| | - Prakash Satwani
- Division of Pediatric Hematology, Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032, USA
| | - Christa Krupski
- Department of Pediatrics, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH 45229-3026, USA
- Cincinnati Children’s Hospital Medical Center, Cancer and Blood Disease Institute, 3333 Burnet Avenue, Cincinnati, OH 45229-3026, USA
| | - Amy Keating
- University of Colorado, Anschutz Medical Campus, Colorado Children’s Hospital, 13123 East 16th Avenue, Aurora, CO 80045, USA
| | - Vanessa Fabrizio
- University of Colorado, Anschutz Medical Campus, Colorado Children’s Hospital, 13123 East 16th Avenue, Aurora, CO 80045, USA
| | - Vasant Chinnabhandar
- Division of Pediatric Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota Medical School, 2450 Riverside Ave S AO-102, Minneapolis, MN 55454, USA
| | - Michael Kunicki
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, 1000 Welch Road, Suite 300, Palo Alto, CA 94304, USA
| | - Kevin J. Curran
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | - Crystal L. Mackall
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, 1000 Welch Road, Suite 300, Palo Alto, CA 94304, USA
- Center for Cancer Cell Therapy, Stanford University School of Medicine, Stanford Cancer Institute, 265 Campus Drive, Stanford, CA 94305, USA
- Division of Blood and Bone Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Room H0101, Stanford, CA 94305-5623, USA
| | - Theodore W. Laetsch
- Department of Pediatrics, Children's Hospital of Philadelphia/University of Pennsylvania, 3401 Civic Center Blvd., Philadelphia, PA 19104, USA
| | - Liora M. Schultz
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, 1000 Welch Road, Suite 300, Palo Alto, CA 94304, USA
| |
Collapse
|
9
|
Roy Moulik N, Keerthivasagam S, Velagala SV, Gollamudi VRM, Agiwale J, Dhamne C, Chichra A, Srinivasan S, Shetty D, Jain H, Subramanian PG, Tembhare P, Chatterjee G, Patkar N, Narula G, Banavali S. Treating relapsed B cell-precursor ALL in children with a setting-adapted mitoxantrone-based intensive chemotherapy protocol (TMH rALL-18 PROTOCOL) - experience from Tata Memorial Hospital, India. Ann Hematol 2023; 102:2835-2844. [PMID: 37479890 DOI: 10.1007/s00277-023-05351-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/30/2023] [Indexed: 07/23/2023]
Abstract
The outlook of relapsed ALL in low- and middle-income countries (LMICs) is dismal due to high treatment-related toxicities and inadequate resources. We report our experience of using a locally adapted mitoxantrone-based protocol for non-high risk (HR) relapsed B-ALL (rALL). A retrospective cum prospective study of standard and intermediate risk (SR and IR) rALL patients treated on TMH rALL-18 protocol (adapted from COG/UKALLR3/Int-Re-ALL protocols) between November 2018 and January 2021 was analyzed. The protocol comprising of 7 blocks of multi-agent chemotherapy including mitoxantrone in induction followed by local irradiation and maintenance, underwent serial modifications based on our experience with initial patients. Eighty-two patients (SR rALL, 3; IR rALL, 79) were treated on TMH rALL-18 protocol. Of 321 grade 3/4 reported toxicities, around 43% (138 toxicities) were noted during induction. Induction chemotherapy was outpatient-based; however, 68 patients (82.9%) required supportive care admissions. Twelve out of 19 patients with gram negative bacilli sepsis (included 7 MDRO) died during reinduction. Five remission deaths were seen during block 3 after which cytarabine was dose reduced (3 g to 2 g/m2). Post-reinduction minimal residual disease was negative in 54 (80.6%) out of 67 evaluable patients. At a median follow-up of 24 months (95% CI 22-27), the estimated 2-year event-free and overall survival of the entire cohort was 58% (95% CI 48.1-69.9) and 60.3% (95% CI 50.5-72). Until the time, targeted therapies are freely accessible in LMICs, strengthening supportive care as well as local adaptation of protocols that strike a fine balance between efficacy and tolerability are mandated.
Collapse
Affiliation(s)
| | | | | | | | - Jayesh Agiwale
- Pediatric Oncology, Tata Memorial Hospital, HBNI, Mumbai, India
| | - Chetan Dhamne
- Pediatric Oncology, Tata Memorial Hospital, HBNI, Mumbai, India
| | | | | | - Dhanlaxmi Shetty
- Cancer Cytogenetics, Tata Memorial Hospital, HBNI, Mumbai, India
| | - Hemani Jain
- Cancer Cytogenetics, Tata Memorial Hospital, HBNI, Mumbai, India
| | | | | | | | - Nikhil Patkar
- Hematopathology, Tata Memorial Hospital, HBNI, Mumbai, India
| | - Gaurav Narula
- Pediatric Oncology, Tata Memorial Hospital, HBNI, Mumbai, India
| | | |
Collapse
|
10
|
Maese LD, Pulsipher MA. Blinatumomab Conundrum in Low-Risk Relapsed B-Cell ALL. J Clin Oncol 2023; 41:4087-4092. [PMID: 37311171 DOI: 10.1200/jco.23.00594] [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: 03/17/2023] [Revised: 04/03/2023] [Accepted: 04/28/2023] [Indexed: 06/15/2023] Open
Abstract
The Oncology Grand Rounds series is designed to place original reports published in the Journal into clinical context. A case presentation is followed by a description of diagnostic and management challenges, a review of the relevant literature, and a summary of the authors' suggested management approaches. The goal of this series is to help readers better understand how to apply the results of key studies, including those published in Journal of Clinical Oncology, to patients seen in their own clinical practice. BACKGROUND The Children's Oncology Group (COG) AALL1331 trial demonstrated improved survival and less toxicity in children with high-/intermediate-risk relapsed ALL receiving blinatumomab compared with intensive chemotherapy before hematopoietic stem-cell transplant (HSCT). The low-risk arm of AALL1331 compared addition of three cycles of blinatumomab to chemotherapy alone, but a survival improvement was not noted. Secondary analyses showed improvement in disease-free survival (DFS) and overall survival (OS) of low-risk patients with bone marrow disease ± extramedullary (EM) involvement (4-year DFS 72.7% ± 5.8% v 53.7% ± 6.7%; 4-year OS 97.1% ± 2.1% v 84.8% ± 4.8%), but failed to show an advantage with blinatumomab for patients with isolated EM relapse. Of note, DFS of isolated CNS (iCNS) relapse was worse than previous studies at 24% on both arms, likely because of decreases in CNS-intensive therapy compared with previous approaches and inadequacy of blinatumomab for controlling CNS disease. CASE Our case of late isolated CNS B-cell ALL relapse outlines challenges for clinicians attempting to decrease toxicity and avoid HSCT: (1) defining of low risk appropriately, (2) attempting to reduce the treatment burden of past protocols, and (3) understanding approach and timing of cranial irradiation. APPROACH Although AALL1331 therapy without blinatumomab leads to excellent survival in patients with isolated testicular relapse, we recommend a modified AALL02P2 backbone of chemotherapy with 1,800 cGy cranial radiotherapy for patients with late iCNS relapse. Future studies integrating chimeric antigen receptor T cells, which have better CNS penetration, may help decrease the intensive treatment burden for patients with late iCNS recurrence.
Collapse
Affiliation(s)
- Luke D Maese
- Intermountain Primary Children's Hospital, Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine at the University of Utah, Salt Lake City, UT
| | - Michael A Pulsipher
- Intermountain Primary Children's Hospital, Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine at the University of Utah, Salt Lake City, UT
| |
Collapse
|
11
|
Hogan LE, Brown PA, Ji L, Xu X, Devidas M, Bhatla T, Borowitz MJ, Raetz EA, Carroll A, Heerema NA, Zugmaier G, Sharon E, Bernhardt MB, Terezakis SA, Gore L, Whitlock JA, Hunger SP, Loh ML. Children's Oncology Group AALL1331: Phase III Trial of Blinatumomab in Children, Adolescents, and Young Adults With Low-Risk B-Cell ALL in First Relapse. J Clin Oncol 2023; 41:4118-4129. [PMID: 37257143 PMCID: PMC10852366 DOI: 10.1200/jco.22.02200] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 02/06/2023] [Accepted: 03/17/2023] [Indexed: 06/02/2023] Open
Abstract
PURPOSE Blinatumomab, a bispecific T-cell engager immunotherapy, is efficacious in relapsed/refractory B-cell ALL (B-ALL) and has a favorable toxicity profile. One aim of the Children's Oncology Group AALL1331 study was to compare survival of patients with low-risk (LR) first relapse of B-ALL treated with chemotherapy alone or chemotherapy plus blinatumomab. PATIENTS AND METHODS After block 1 reinduction, patients age 1-30 years with LR first relapse of B-ALL were randomly assigned to block 2/block 3/two continuation chemotherapy cycles/maintenance (arm C) or block 2/two cycles of continuation chemotherapy intercalated with three blinatumomab blocks/maintenance (arm D). Patients with CNS leukemia received 18 Gy cranial radiation during maintenance and intensified intrathecal chemotherapy. The primary and secondary end points were disease-free survival (DFS) and overall survival (OS). RESULTS The 4-year DFS/OS for the 255 LR patients accrued between December 2014 and September 2019 were 61.2% ± 5.0%/90.4% ± 3.0% for blinatumomab versus 49.5% ± 5.2%/79.6% ± 4.3% for chemotherapy (P = .089/P = .11). For bone marrow (BM) ± extramedullary (EM) (BM ± EM; n = 174) relapses, 4-year DFS/OS were 72.7% ± 5.8%/97.1% ± 2.1% for blinatumomab versus 53.7% ± 6.7%/84.8% ± 4.8% for chemotherapy (P = .015/P = .020). For isolated EM (IEM; n = 81) relapses, 4-year DFS/OS were 36.6% ± 8.2%/76.5% ± 7.5% for blinatumomab versus 38.8% ± 8.0%/68.8% ± 8.6% for chemotherapy (P = .62/P = .53). Blinatumomab was well tolerated and patients had low adverse event rates. CONCLUSION For children, adolescents, and young adults with B-ALL in LR first relapse, there was no statistically significant difference in DFS or OS between the blinatumomab and standard chemotherapy arms overall. However, blinatumomab significantly improved DFS and OS for the two thirds of patients with BM ± EM relapse, establishing a new standard of care for this population. By contrast, similar outcomes and poor DFS for both arms were observed in the one third of patients with IEM; new treatment approaches are needed for these patients (ClinicalTrials.gov identifier: NCT02101853).
Collapse
Affiliation(s)
- Laura E. Hogan
- Department of Pediatrics, Stony Brook Children's, Stony Brook, NY
| | | | - Lingyun Ji
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Xinxin Xu
- Children's Oncology Group, Monrovia, CA
| | - Meenakshi Devidas
- Department of Global Pediatric Medicine, St Jude Children's Research Hospital, Memphis, TN
| | - Teena Bhatla
- Childrens Hospital of New Jersey at Newark Beth Israel, Newark, NJ
| | - Michael J. Borowitz
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | | - Nyla A. Heerema
- Department of Pathology, The Ohio State University, Columbus, OH
| | | | - Elad Sharon
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Cancer Therapy Evaluation Program, Bethesda, MD
| | - Melanie B. Bernhardt
- Section of Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | | | - Lia Gore
- University of Colorado School of Medicine and Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO
| | - James A. Whitlock
- Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - 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, PA
| | - Mignon L. Loh
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA
- Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA
| |
Collapse
|
12
|
Harris RD, Bernhardt MB, Zobeck M, Taylor O, Gramatges MM, Schafer ES, Lupo PJ, Rabin KR, Scheurer ME, Brown AL. Ethnic-specific predictors of neurotoxicity among patients with pediatric acute lymphoblastic leukemia after high-dose methotrexate. Cancer 2023; 129:1287-1294. [PMID: 36692972 PMCID: PMC10625847 DOI: 10.1002/cncr.34646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/02/2022] [Accepted: 12/10/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND High-dose methotrexate (HD-MTX; 5000 mg/m2 ) is an important component of curative therapy in many treatment regimens for high-risk pediatric acute lymphoblastic leukemia (ALL). However, methotrexate therapy can result in dose-limiting neurotoxicity, which may disproportionately affect Latino children. This study evaluated risk factors for neurotoxicity after HD-MTX in an ethnically diverse population of patients with ALL. METHODS The authors retrospectively reviewed the medical records of patients who were diagnosed with ALL and treated with HD-MTX at Texas Children's Cancer Center (2010-2017). Methotrexate neurotoxicity was defined as a neurologic episode (e.g., seizures or stroke-like symptoms) occurring within 21 days of HD-MTX that resulted in methotrexate treatment modifications. Mixed effects multivariable logistic regression was used to estimate the odds ratio (OR) and corresponding 95% confidence interval (CI) for the association between clinical factors and neurotoxicity. RESULTS Overall, 351 patients (58.1% Latino) who received 1183 HD-MTX infusions were evaluated. Thirty-five patients (10%) experienced neurotoxicity, 71% of whom were Latino. After adjusting for clinical risk factors, the authors observed that serum creatinine elevations ≥50% of baseline were associated with a three-fold increased odds (OR, 3.32; 95% CI, 0.98-11.21; p = .05) for neurotoxicity compared with creatinine elevation <25%. Notably, predictors of neurotoxicity differed by ethnicity. Specifically, Latino children experienced a nearly six-fold increase in neurotoxicity odds (OR, 5.80; 95% CI, 1.39-24.17; p = .02) with serum creatinine elevation ≥50% compared with creatinine elevation <25%. CONCLUSIONS The current findings indicate that serum creatinine elevations ≥50% may be associated with an increased risk for neurotoxicity among Latino children with ALL and may identify potential candidates for therapeutic or supportive care interventions.
Collapse
Affiliation(s)
- Rachel D. Harris
- Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas
- Texas Children’s Hospital, Texas Children’s Cancer and Hematology Centers, Houston, Texas
| | - M. Brooke Bernhardt
- Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas
- Texas Children’s Hospital, Texas Children’s Cancer and Hematology Centers, Houston, Texas
| | - Mark Zobeck
- Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas
- Texas Children’s Hospital, Texas Children’s Cancer and Hematology Centers, Houston, Texas
| | - Olga Taylor
- Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas
- Texas Children’s Hospital, Texas Children’s Cancer and Hematology Centers, Houston, Texas
| | - M. Monica Gramatges
- Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas
- Texas Children’s Hospital, Texas Children’s Cancer and Hematology Centers, Houston, Texas
| | - Eric S. Schafer
- Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas
- Texas Children’s Hospital, Texas Children’s Cancer and Hematology Centers, Houston, Texas
| | - Philip J. Lupo
- Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas
- Texas Children’s Hospital, Texas Children’s Cancer and Hematology Centers, Houston, Texas
| | - Karen R. Rabin
- Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas
- Texas Children’s Hospital, Texas Children’s Cancer and Hematology Centers, Houston, Texas
| | - Michael E. Scheurer
- Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas
- Texas Children’s Hospital, Texas Children’s Cancer and Hematology Centers, Houston, Texas
| | - Austin L. Brown
- Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas
- Texas Children’s Hospital, Texas Children’s Cancer and Hematology Centers, Houston, Texas
| |
Collapse
|
13
|
Kopmar NE, Cassaday RD. How I prevent and treat central nervous system disease in adults with acute lymphoblastic leukemia. Blood 2023; 141:1379-1388. [PMID: 36548957 PMCID: PMC10082377 DOI: 10.1182/blood.2022017035] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/28/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
The central nervous system (CNS) is the most important site of extramedullary disease in adults with acute lymphoblastic leukemia (ALL). Although CNS disease is identified only in a minority of patients at the time of diagnosis, subsequent CNS relapses (either isolated or concurrent with other sites) occur in some patients even after the delivery of prophylactic therapy targeted to the CNS. Historically, prophylaxis against CNS disease has included intrathecal (IT) chemotherapy and radiotherapy (RT), although the latter is being used with decreasing frequency. Treatment of a CNS relapse usually involves intensive systemic therapy and cranial or craniospinal RT along with IT therapy and consideration of allogeneic hematopoietic cell transplant. However, short- and long-term toxicities can make these interventions prohibitively risky, particularly for older adults. As new antibody-based immunotherapy agents have been approved for relapsed/refractory B-cell ALL, their use specifically for patients with CNS disease is an area of keen interest not only because of the potential for efficacy but also concerns of unique toxicity to the CNS. In this review, we discuss data-driven approaches for these common and challenging clinical scenarios as well as highlight how recent findings potentially support the use of novel immunotherapeutic strategies for CNS disease.
Collapse
Affiliation(s)
- Noam E. Kopmar
- Division of Hematology, Department of Medicine, University of Washington School of Medicine, University of Washington, Seattle, WA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Ryan D. Cassaday
- Division of Hematology, Department of Medicine, University of Washington School of Medicine, University of Washington, Seattle, WA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| |
Collapse
|
14
|
Development of a Therapeutic Drug Monitoring Strategy for the Optimization of Vincristine Treatment in Pediatric Oncology Populations in Africa. Ther Drug Monit 2023; 45:354-363. [PMID: 36917736 DOI: 10.1097/ftd.0000000000001090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
BACKGROUND Recent studies have reported ethnic differences in vincristine exposure and outcomes such as toxicity. This resulted in the hypothesis of subtherapeutic dosing in African children. To optimize individual treatment, a strategy to identify subtherapeutic exposure using therapeutic drug monitoring is essential. The aim of the current study was to develop a strategy for therapeutic drug monitoring of vincristine in African children to meet the following criteria: (1) identify patients with low vincristine exposure with sufficient sensitivity (>70%), (2) determine vincristine exposure with a limited sampling strategy design of 3 samples, and (3) allow all samples to be collected within 4 hours after administration. METHODS An in silico simulation study was performed using a previously described population pharmacokinetic model and real-life demographic dataset of Kenyan and Malawian pediatric oncology patients. Two different therapeutic drug monitoring strategies were evaluated: (1) Bayesian approach and (2) pharmacometric nomogram. The sampling design was optimized using the constraints described above. Sensitivity analysis was performed to investigate the influence of missing samples, erroneous sampling times, and different boundaries on the nomogram weight bands. RESULTS With the Bayesian approach, 43.3% of the estimated individual exposure values had a prediction error of ≥20% owing to extremely high shrinkage. The Bayesian approach did not improve with alternative sampling designs within sampling constraints. However, the pharmacometric nomogram could identify patients with low vincristine exposure with a sensitivity, specificity, and accuracy of 75.1%, 76.4%, and 75.9%, respectively. The pharmacometric nomogram performed similarly for different weight bands. CONCLUSIONS The pharmacometric nomogram was able to identify patients with low vincristine exposure with high sensitivity, with 3 blood samples collected at 1, 1.5, and 4 hours after administration. Missing samples should be avoided, and the 3 scheduled samples should be collected within 15, 5, and 15 minutes of 1, 1.5, and 4 hours after administration, respectively.
Collapse
|
15
|
Wu CY, Li GT, Chu CC, Guo HL, Fang WR, Li T, Wang YR, Xu J, Hu YH, Zhou L, Chen F. Proactive therapeutic drug monitoring of vincristine in pediatric and adult cancer patients: current supporting evidence and future efforts. Arch Toxicol 2023; 97:377-392. [PMID: 36418572 DOI: 10.1007/s00204-022-03418-8] [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: 10/08/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022]
Abstract
Vincristine (VCR), an effective antitumor drug, has been utilized in several polytherapy regimens for acute lymphoblastic leukemia, neuroblastoma and rhabdomyosarcoma. However, clinical evidence shows that the metabolism of VCR varies greatly among patients. The traditional based body surface area (BSA) administration method is prone to insufficient exposure to VCR or severe VCR-induced peripheral neurotoxicity (VIPN). Therefore, reliable strategies are urgently needed to improve efficacy and reduce VIPN. Due to the unpredictable pharmacokinetic changes of VCR, therapeutic drug monitoring (TDM) may help to ensure its efficacy and to manage VIPN. At present, there is a lot of supporting evidence for the suitability of applying TDM to VCR therapy. Based on the consensus guidelines drafted by the International Association of Therapeutic Drug Monitoring and Clinical Toxicology (IATDMCT), this review aimed to summarize various available data to evaluate the potential utility of VCR TDM for cancer patients. Of note, valuable evidence has accumulated on pharmacokinetics variability, pharmacodynamics, drug exposure-clinical response relationship, biomarkers for VIPN prediction, and assays for VCR monitoring. However, there are still many relevant clinical pharmacological questions that cannot yet be answered merely based on insufficient evidence. Currently, we cannot recommend a therapeutic exposure range and cannot yet provide a dose-adaptation strategy for clinicians and patients. In areas where the evidence is not yet sufficient, more research is needed in the future. The precision medicine of VCR cannot rely on TDM alone and needs to consider the clinical, environmental, genetic background and patient-specific factors as a whole.
Collapse
Affiliation(s)
- Chun-Ying Wu
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China.,School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Guan-Ting Li
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Chen-Chao Chu
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China.,School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hong-Li Guo
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Wei-Rong Fang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Tao Li
- Department of Solid Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yong-Ren Wang
- Department of Hematology /Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Xu
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Ya-Hui Hu
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China.
| | - Li Zhou
- Department of Hematology /Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China.
| | - Feng Chen
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China.
| |
Collapse
|
16
|
Thastrup M, Duguid A, Mirian C, Schmiegelow K, Halsey C. Central nervous system involvement in childhood acute lymphoblastic leukemia: challenges and solutions. Leukemia 2022; 36:2751-2768. [PMID: 36266325 PMCID: PMC9712093 DOI: 10.1038/s41375-022-01714-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 09/17/2022] [Accepted: 09/22/2022] [Indexed: 11/10/2022]
Abstract
Delivery of effective anti-leukemic agents to the central nervous system (CNS) is considered essential for cure of childhood acute lymphoblastic leukemia. Current CNS-directed therapy comprises systemic therapy with good CNS-penetration accompanied by repeated intrathecal treatments up to 26 times over 2-3 years. This approach prevents most CNS relapses, but is associated with significant short and long term neurotoxicity. Despite this burdensome therapy, there have been no new drugs licensed for CNS-leukemia since the 1960s, when very limited anti-leukemic agents were available and there was no mechanistic understanding of leukemia survival in the CNS. Another major barrier to improved treatment is that we cannot accurately identify children at risk of CNS relapse, or monitor response to treatment, due to a lack of sensitive biomarkers. A paradigm shift in treating the CNS is needed. The challenges are clear - we cannot measure CNS leukemic load, trials have been unable to establish the most effective CNS treatment regimens, and non-toxic approaches for relapsed, refractory, or intolerant patients are lacking. In this review we discuss these challenges and highlight research advances aiming to provide solutions. Unlocking the potential of risk-adapted non-toxic CNS-directed therapy requires; (1) discovery of robust diagnostic, prognostic and response biomarkers for CNS-leukemia, (2) identification of novel therapeutic targets combined with associated investment in drug development and early-phase trials and (3) engineering of immunotherapies to overcome the unique challenges of the CNS microenvironment. Fortunately, research into CNS-ALL is now making progress in addressing these unmet needs: biomarkers, such as CSF-flow cytometry, are now being tested in prospective trials, novel drugs are being tested in Phase I/II trials, and immunotherapies are increasingly available to patients with CNS relapses. The future is hopeful for improved management of the CNS over the next decade.
Collapse
Affiliation(s)
- Maria Thastrup
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Alasdair Duguid
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Christian Mirian
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Proteomics Program, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christina Halsey
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
17
|
Chotsampancharoen T, Songthawee N, Chavananon S, Sripornsawan P, McNeil EB. Relapsed Childhood Acute Lymphoblastic Leukemia: Experience from a Single Tertiary Center in Thailand. Asian Pac J Cancer Prev 2022; 23:3517-3522. [PMID: 36308378 PMCID: PMC9924329 DOI: 10.31557/apjcp.2022.23.10.3517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND The outcomes of relapsed childhood acute lymphoblastic leukemia (ALL) in developed countries have improved over time as a result of risk-adapted, minimal residual disease-directed therapy, hematopoietic stem cell transplantation, and immunotherapy. There are few studies that have examined survival in relapsed childhood ALL in resource-limited countries. Therefore, this study aimed to assess the prognostic factors and survival outcome of relapsed childhood ALL in a major tertiary center in Southern Thailand. METHODS The medical records of patients with ALL aged <15 years between January 2000 and December 2019 were retrospectively reviewed. The Kaplan-Meier method was used to depict the overall survival (OS). RESULTS A total of 472 patients with ALL were enrolled and relapsed ALL was found in 155 (32.8%) patients. Of these, 131 (84.5%) and 24 (15.5%) had B-cell and T-cell phenotypes, respectively. One hundred thirteen (72.9%) and 42 (27.1%) patients had early and late relapses, respectively. The most common site of relapse was bone marrow in 102 patients (65.8%). One hundred twenty-eight (82.6%) patients received treatment while 27 (17.4%) patients refused treatment. The 5-year OS of all relapsed patients was 11.9%. The 5-year OS among the patients with early relapse was significantly lower than in the patients with late relapse (5.3% vs. 29.1%, respectively, p <0.0001). Site and immunophenotype were not associated with survival of relapsed ALL. The median survival times among the patients who received and refused relapse chemotherapy were 11.8 and 3.1 months, respectively (p <0.0001). CONCLUSION The relapse rate accounted for one third of patients with ALL with the 5-year OS of 12%. Early relapse and those who refused treatment were associated with poor survival outcome.
Collapse
Affiliation(s)
- Thirachit Chotsampancharoen
- Division of Hematology and Oncology, Department of Pediatrics, Prince of Songkla University, Hat Yai, Thailand. ,For Correspondence:
| | - Natsaruth Songthawee
- Division of Hematology and Oncology, Department of Pediatrics, Prince of Songkla University, Hat Yai, Thailand.
| | - Shevachut Chavananon
- Division of Hematology and Oncology, Department of Pediatrics, Prince of Songkla University, Hat Yai, Thailand.
| | - Pornpun Sripornsawan
- Division of Hematology and Oncology, Department of Pediatrics, Prince of Songkla University, Hat Yai, Thailand.
| | - Edward B. McNeil
- Epidemiology Unit, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand.
| |
Collapse
|
18
|
Jensen KS, Oskarsson T, Lähteenmäki PM, Flaegstad T, Jónsson ÓG, Svenberg P, Schmiegelow K, Heyman M, Norén-Nyström U, Schrøder H, Albertsen BK. Temporal changes in incidence of relapse and outcome after relapse of childhood acute lymphoblastic leukemia over three decades; a Nordic population-based cohort study. Leukemia 2022; 36:1274-1282. [PMID: 35314777 DOI: 10.1038/s41375-022-01540-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/11/2022] [Accepted: 03/01/2022] [Indexed: 11/09/2022]
Abstract
Relapse remains the main obstacle to curing childhood acute lymphoblastic leukemia (ALL). The aims of this study were to compare incidence of relapse, prognostic factors, and survival after relapse between three consecutive Nordic Society of Pediatric Hematology and Oncology trials. Relapse occurred as a primary event in 638 of 4 458 children (1.0-14.9 years) diagnosed with Ph-negative ALL between 1992 and 2018. The 5-year cumulative incidence of relapse was 17.3% (95% CI 15.4-19.2%) and 16.5% (95% CI 14.3-18.8%) for patients in the ALL1992 and ALL2000 trials, respectively, but decreased to 8.4% (95% CI 7.0-10.1%) for patients in the ALL2008 trial. No changes in duration of first complete remission and site of relapse were observed over time; however, high hyperdiploidy, and t(12;21) decreased in the ALL2008 trial. The 4-year overall survival after relapse was 56.6% (95% CI 52.5-60.5%) and no statistically significant temporal improvements were observed. Age ≥10 years, T-cell immunophenotype, bone-marrow involvement, early and very early relapse, hypodiploidy, and Down syndrome all independently predicted worse outcome after relapse. Improvements in the primary treatment of childhood ALL has resulted in fewer relapses. However, failure to improve outcome of remaining relapses suggests a selection of harder-to-cure relapses and calls for new therapeutic strategies.
Collapse
Affiliation(s)
- Karen Schow Jensen
- Department of Paediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Trausti Oskarsson
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden.,Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Päivi M Lähteenmäki
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden.,Department of Pediatric and Adolescent Hematology/Oncology, Turku University Hospital, FICAN-west, and Turku University, Turku, Finland
| | - Trond Flaegstad
- Department of Pediatrics, Institute of Clinical Medicine, University of Tromsø, Tromsø, Norway.,Department of Pediatrics, University Hospital of North Norway, Tromsø, Norway
| | | | - Petter Svenberg
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mats Heyman
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | | | - Henrik Schrøder
- Department of Paediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Birgitte Klug Albertsen
- Department of Paediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark. .,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
19
|
Fabrizio VA, Phillips CL, Lane A, Baggott C, Prabhu S, Egeler E, Mavroukakis S, Pacenta H, Rossoff J, Stefanski HE, Talano JA, Moskop A, Margossian SP, Verneris MR, Myers GD, Karras NA, Brown PA, Qayed M, Hermiston M, Satwani P, Krupski C, Keating AK, Wilcox R, Rabik CA, Chinnabhandar V, Kunicki M, Goksenin AY, Curran KJ, Mackall CL, Laetsch TW, Schultz LM. Tisagenlecleucel outcomes in relapsed/refractory extramedullary ALL: a Pediatric Real World CAR Consortium Report. Blood Adv 2022; 6:600-610. [PMID: 34794180 PMCID: PMC8791593 DOI: 10.1182/bloodadvances.2021005564] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 10/27/2021] [Indexed: 11/20/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells have transformed the therapeutic options for relapsed/refractory (R/R) B-cell acute lymphoblastic leukemia. Data for CAR therapy in extramedullary (EM) involvement are limited. Retrospective data were abstracted from the Pediatric Real World CAR Consortium (PRWCC) of 184 infused patients from 15 US institutions. Response (complete response) rate, overall survival (OS), relapse-free survival (RFS), and duration of B-cell aplasia (BCA) in patients referred for tisagenlecleucel with EM disease (both central nervous system (CNS)3 and non-CNS EM) were compared with bone marrow (BM) only. Patients with CNS disease were further stratified for comparison. Outcomes are reported on 55 patients with EM disease before CAR therapy (CNS3, n = 40; non-CNS EM, n = 15). The median age at infusion in the CNS cohort was 10 years (range, <1-25 years), and in the non-CNS EM cohort it was 13 years (range, 2-26 years). In patients with CNS disease, 88% (35 of 40) achieved a complete response vs only 66% (10 of 15) with non-CNS EM disease. Patients with CNS disease (both with and without BM involvement) had 24-month OS outcomes comparable to those of non-CNS EM or BM only (P = .41). There was no difference in 12-month RFS between CNS, non-CNS EM, or BM-only patients (P = .92). No increased toxicity was seen with CNS or non-CNS EM disease (P = .3). Active CNS disease at time of infusion did not affect outcomes. Isolated CNS disease trended toward improved OS compared with combined CNS and BM (P = .12). R/R EM disease can be effectively treated with tisagenlecleucel; toxicity, relapse, and survival rates are comparable to those of patients with BM-only disease. Outcomes for isolated CNS relapse are encouraging.
Collapse
Affiliation(s)
- Vanessa A Fabrizio
- University of Colorado, Anschutz Medical Campus, Colorado Children's Hospital, Aurora, CO
| | - Christine L Phillips
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH
- Cincinnati Children's Hospital Medical Center, Cancer and Blood Diseases Institute, Cincinnati, OH
| | - Adam Lane
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH
| | - Christina Baggott
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Snehit Prabhu
- Stanford University School of Medicine, Stanford Cancer Institute, Center for Cancer Cell Therapy, Stanford, CA
| | - Emily Egeler
- Stanford University School of Medicine, Stanford Cancer Institute, Center for Cancer Cell Therapy, Stanford, CA
| | - Sharon Mavroukakis
- Stanford University School of Medicine, Stanford Cancer Institute, Center for Cancer Cell Therapy, Stanford, CA
| | - Holly Pacenta
- Department of Pediatrics, The University of Texas Southwestern Medical Center/Children's Health, Dallas, TX
| | - Jenna Rossoff
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL
| | - Heather E Stefanski
- Department of Pediatrics, Division of Pediatric Blood and Marrow Transplantation, University of Minnesota Medical School, Minneapolis, MN
| | - Julie-An Talano
- Department of Pediatric Hematology Oncology, Medical College of Wisconsin, Milwaukee, WI
| | - Amy Moskop
- Department of Pediatric Hematology Oncology, Medical College of Wisconsin, Milwaukee, WI
| | - Steven P Margossian
- Harvard Medical School, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Pediatric Hematology-Oncology, Boston, MA
| | - Michael R Verneris
- University of Colorado, Anschutz Medical Campus, Colorado Children's Hospital, Aurora, CO
| | | | - Nicole A Karras
- Department of Pediatrics, City of Hope National Medical Center, Duarte, CA
| | - Patrick A Brown
- Department of Oncology, Sidney Kimmel Cancer Center at John Hopkins School of Medicine, Baltimore, MD
| | - Muna Qayed
- Emory University and Children's Healthcare of Atlanta, Atlanta, GA
| | - Michelle Hermiston
- Benioff Children's Hospital, University of California San Francisco, San Francisco, CA
| | - Prakash Satwani
- Division of Pediatric Hematology, Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Medical Center, New York, NY
| | - Christa Krupski
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH
- Cincinnati Children's Hospital Medical Center, Cancer and Blood Diseases Institute, Cincinnati, OH
| | - Amy K Keating
- University of Colorado, Anschutz Medical Campus, Colorado Children's Hospital, Aurora, CO
| | | | - Cara A Rabik
- Department of Oncology, Sidney Kimmel Cancer Center at John Hopkins School of Medicine, Baltimore, MD
| | - Vasant Chinnabhandar
- Department of Pediatrics, Division of Pediatric Blood and Marrow Transplantation, University of Minnesota Medical School, Minneapolis, MN
| | - Michael Kunicki
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - A Yasemin Goksenin
- Benioff Children's Hospital, University of California San Francisco, San Francisco, CA
| | - Kevin J Curran
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medical College, New York, NY
| | - Crystal L Mackall
- Division of Hematology and Oncology, Department of Pediatrics, Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, CA
- Division of Stem Cell Transplantation and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Theodore W Laetsch
- Department of Pediatrics, The University of Texas Southwestern Medical Center/Children's Health, Dallas, TX
- Department of Pediatrics and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA; and
| | - Liora M Schultz
- Department of Pediatrics, Division of Hematology and Oncology, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
20
|
Mengxuan S, Fen Z, Runming J. Novel Treatments for Pediatric Relapsed or Refractory Acute B-Cell Lineage Lymphoblastic Leukemia: Precision Medicine Era. Front Pediatr 2022; 10:923419. [PMID: 35813376 PMCID: PMC9259965 DOI: 10.3389/fped.2022.923419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/02/2022] [Indexed: 12/05/2022] Open
Abstract
With the markedly increased cure rate for children with newly diagnosed pediatric B-cell acute lymphoblastic leukemia (B-ALL), relapse and refractory B-ALL (R/R B-ALL) remain the primary cause of death worldwide due to the limitations of multidrug chemotherapy. As we now have a more profound understanding of R/R ALL, including the mechanism of recurrence and drug resistance, prognostic indicators, genotypic changes and so on, we can use newly emerging technologies to identify operational molecular targets and find sensitive drugs for individualized treatment. In addition, more promising and innovative immunotherapies and molecular targeted drugs that are expected to kill leukemic cells more effectively while maintaining low toxicity to achieve minimal residual disease (MRD) negativity and better bridge hematopoietic stem cell transplantation (HSCT) have also been widely developed. To date, the prognosis of pediatric patients with R/R B-ALL has been enhanced markedly thanks to the development of novel drugs. This article reviews the new advancements of several promising strategies for pediatric R/R B-ALL.
Collapse
Affiliation(s)
- Shang Mengxuan
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhou Fen
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin Runming
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Harada T, Toyoda H, Tsuboya N, Hanaki R, Amano K, Hirayama M. Successful hematopoietic stem cell transplantation for two patients with relapse of intrachromosomal amplification of chromosome 21-positive B-cell precursor acute lymphoblastic leukemia. Front Pediatr 2022; 10:960126. [PMID: 36160794 PMCID: PMC9492991 DOI: 10.3389/fped.2022.960126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/09/2022] [Indexed: 11/30/2022] Open
Abstract
In children with relapsed acute lymphoblastic leukemia (ALL), it is essential to identify patients in need of treatment intensification. Minimal residual disease (MRD)-based treatment stratification resulted in excellent survival in children with late relapsed B-cell precursor (BCP)-ALL. Chemotherapy alone produced a favorable outcome in patients with negative MRD after induction. The genetic abnormality also plays an important role in determining the prognosis and stratification for treatment. Intrachromosomal amplification of chromosome 21 (iAMP21) is associated with a poor outcome and a high risk for relapse, and there is no standard treatment after relapse. Herein, we present two patients with relapsed iAMP21-positive ALL who were successfully treated by cord blood transplantation (CBT). Although both patients had late bone marrow relapse and favorable MRD response, CBT was performed due to iAMP21 positive. Patients 1 and 2 have been in remission post-CBT for 15 and 45 months, respectively. Patients with relapsed iAMP21-positive ALL may be considered for stem cell transplantation even in late relapses and favorable MRD response.
Collapse
Affiliation(s)
- Tomoya Harada
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hidemi Toyoda
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Japan
| | - Naoki Tsuboya
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Japan
| | - Ryo Hanaki
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Japan
| | - Keishiro Amano
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Japan
| | - Masahiro Hirayama
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
22
|
Truong TH, Jinca C, Mann G, Arghirescu S, Buechner J, Merli P, Whitlock JA. Allogeneic Hematopoietic Stem Cell Transplantation for Children With Acute Lymphoblastic Leukemia: Shifting Indications in the Era of Immunotherapy. Front Pediatr 2021; 9:782785. [PMID: 35004545 PMCID: PMC8733383 DOI: 10.3389/fped.2021.782785] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/08/2021] [Indexed: 12/26/2022] Open
Abstract
Pediatric acute lymphoblastic leukemia generally carries a good prognosis, and most children will be cured and become long-term survivors. However, a portion of children will harbor high-risk features at the time of diagnosis, have a poor response to upfront therapy, or suffer relapse necessitating more intensive therapy, which may include allogeneic hematopoietic stem cell transplant (HSCT). Recent advances in risk stratification, improved detection and incorporation of minimal residual disease (MRD), and intensification of upfront treatment have changed the indications for HSCT over time. For children in first complete remission, HSCT is generally reserved for those with the highest risk of relapse. These include patients with unfavorable features/cytogenetics who also have a poor response to induction and consolidation chemotherapy, usually reflected by residual blasts after prednisone or by detectable MRD at pre-defined time points. In the relapsed setting, children with first relapse of B-cell ALL are further stratified for HSCT depending on the time and site of relapse, while all patients with T-cell ALL are generally consolidated with HSCT. Alternatives to HSCT have also emerged over the last decade including immunotherapy and chimeric antigen receptor (CAR) T-cell therapy. These novel agents may spare toxicity while attempting to achieve MRD-negative remission in the most refractory cases and serve as a bridge to HSCT. In some situations, these emerging therapies can indeed be curative for some children with relapsed or resistant disease, thus, obviating the need for HSCT. In this review, we seek to summarize the role of HSCT in the current era of immunotherapy.
Collapse
Affiliation(s)
- Tony H. Truong
- Division of Pediatric Oncology, Blood and Marrow Transplant/Cellular Therapy, Alberta Children's Hospital, University of Calgary, Calgary, AB, Canada
| | - Cristian Jinca
- Department of Pediatrics, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| | - Georg Mann
- Children's Cancer Research Institute, St. Anna Children's Hospital, Vienna, Austria
| | - Smaranda Arghirescu
- Department of Pediatrics, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| | - Jochen Buechner
- Department of Pediatric Hematology and Oncology, Oslo University Hospital, Oslo, Norway
| | - Pietro Merli
- Department of Pediatric Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - James A. Whitlock
- Department of Paediatrics, Hospital for Sick Children/University of Toronto, Toronto, ON, Canada
| |
Collapse
|
23
|
Ceppi F, Rizzati F, Colombini A, Conter V, Cazzaniga G. Utilizing the prognostic impact of minimal residual disease in treatment decisions for pediatric acute lymphoblastic leukemia. Expert Rev Hematol 2021; 14:795-807. [PMID: 34374613 DOI: 10.1080/17474086.2021.1967137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Acute lymphoblastic leukemia (ALL) is the first pediatric cancer where the assessment of early response to therapy by minimal residual disease (MRD) monitoring has demonstrated its importance to improve risk-based treatment approaches. The most standardized tools to study MRD in ALL are multiparametric flow cytometry and realtime-quantitative polymerase chain reaction amplification-based methods. In recent years, MRD measurement has reached greater levels of sensitivity and standardization through international laboratory networks collaboration. AREAS COVERED We herewith describe how to assess and apply the prognostic impact of MRD in treatment decisions, with specific focus on pediatric ALL. We also highlight the role of MRD monitoring in the context of genetically homogeneous subgroups of pediatric ALL. However, some queries remain to be addressed and emerging technologies hold the promise of improving MRD detection in ALL patients. EXPERT OPINION Emerging technologies, like next generation flow cytometry, droplet digital PCR, and next generation sequencing appear to be important methods for assessing MRD in pediatric ALL. These more specific and/or sensitive MRD monitoring methods may help to predict relapse with greater accuracy, and are currently being used in clinical trials to improve pediatric ALL outcome by optimizing patient stratification and earlier MRD-based interventional therapy.
Collapse
Affiliation(s)
- Francesco Ceppi
- Pediatric Hematology-Oncology Unit, Division of Pediatrics, Woman-Mother-Child Department, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Frida Rizzati
- Pediatric Hematology-Oncology Unit, Division of Pediatrics, Woman-Mother-Child Department, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Antonella Colombini
- Pediatric Hematology-Oncology, University Milano Bicocca, Fondazione MBBM/Ospedale San Gerardo, Monza, Italy
| | - Valentino Conter
- Pediatric Hematology-Oncology, University Milano Bicocca, Fondazione MBBM/Ospedale San Gerardo, Monza, Italy
| | - Giovanni Cazzaniga
- Centro Ricerca Tettamanti, Pediatrics, School of Medicine, University of Milano Bicocca, Fondazione MBBM/Ospedale San Gerardo, Monza, Italy.,Medical Genetics, School of Medicine, University of Milano Bicocca, Monza, Italy
| |
Collapse
|
24
|
Brown PA, Ji L, Xu X, Devidas M, Hogan LE, Borowitz MJ, Raetz EA, Zugmaier G, Sharon E, Bernhardt MB, Terezakis SA, Gore L, Whitlock JA, Pulsipher MA, Hunger SP, Loh ML. Effect of Postreinduction Therapy Consolidation With Blinatumomab vs Chemotherapy on Disease-Free Survival in Children, Adolescents, and Young Adults With First Relapse of B-Cell Acute Lymphoblastic Leukemia: A Randomized Clinical Trial. JAMA 2021; 325:833-842. [PMID: 33651090 PMCID: PMC7926290 DOI: 10.1001/jama.2021.0669] [Citation(s) in RCA: 218] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
IMPORTANCE Standard chemotherapy for first relapse of B-cell acute lymphoblastic leukemia (B-ALL) in children, adolescents, and young adults is associated with high rates of severe toxicities, subsequent relapse, and death, especially for patients with early relapse (high risk) or late relapse with residual disease after reinduction chemotherapy (intermediate risk). Blinatumomab, a bispecific CD3 to CD19 T cell-engaging antibody construct, is efficacious in relapsed/refractory B-ALL and has a favorable toxicity profile. OBJECTIVE To determine whether substituting blinatumomab for intensive chemotherapy in consolidation therapy would improve survival in children, adolescents, and young adults with high- and intermediate-risk first relapse of B-ALL. DESIGN, SETTING, AND PARTICIPANTS This trial was a randomized phase 3 clinical trial conducted by the Children's Oncology Group at 155 hospitals in the US, Canada, Australia, and New Zealand with enrollment from December 2014 to September 2019 and follow-up until September 30, 2020. Eligible patients included those aged 1 to 30 years with B-ALL first relapse, excluding those with Down syndrome, Philadelphia chromosome-positive ALL, prior hematopoietic stem cell transplant, or prior blinatumomab treatment (n = 669). INTERVENTIONS All patients received a 4-week reinduction chemotherapy course, followed by randomized assignment to receive 2 cycles of blinatumomab (n = 105) or 2 cycles of multiagent chemotherapy (n = 103), each followed by transplant. MAIN OUTCOME AND MEASURES The primary end point was disease-free survival and the secondary end point was overall survival, both from the time of randomization. The threshold for statistical significance was set at a 1-sided P <.025. RESULTS Among 208 randomized patients (median age, 9 years; 97 [47%] females), 118 (57%) completed the randomized therapy. Randomization was terminated at the recommendation of the data and safety monitoring committee without meeting stopping rules for efficacy or futility; at that point, 80 of 131 planned events occurred. With 2.9 years of median follow-up, 2-year disease-free survival was 54.4% for the blinatumomab group vs 39.0% for the chemotherapy group (hazard ratio for disease progression or mortality, 0.70 [95% CI, 0.47-1.03]); 1-sided P = .03). Two-year overall survival was 71.3% for the blinatumomab group vs 58.4% for the chemotherapy group (hazard ratio for mortality, 0.62 [95% CI, 0.39-0.98]; 1-sided P = .02). Rates of notable serious adverse events included infection (15%), febrile neutropenia (5%), sepsis (2%), and mucositis (1%) for the blinatumomab group and infection (65%), febrile neutropenia (58%), sepsis (27%), and mucositis (28%) for the chemotherapy group. CONCLUSIONS AND RELEVANCE Among children, adolescents, and young adults with high- and intermediate-risk first relapse of B-ALL, postreinduction treatment with blinatumomab compared with chemotherapy, followed by transplant, did not result in a statistically significant difference in disease-free survival. However, study interpretation is limited by early termination with possible underpowering for the primary end point. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02101853.
Collapse
Affiliation(s)
- Patrick A. Brown
- Departments of Oncology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lingyun Ji
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles
| | - Xinxin Xu
- Children's Oncology Group, Monrovia, California
| | - Meenakshi Devidas
- Department of Global Pediatric Medicine, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Laura E. Hogan
- Department of Pediatrics, Stony Brook Children’s, Stony Brook, New York
| | - Michael J. Borowitz
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | | | - Elad Sharon
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Cancer Therapy Evaluation Program, Bethesda, Maryland
| | - Melanie B. Bernhardt
- Section of Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | | | - Lia Gore
- University of Colorado School of Medicine and Center for Cancer and Blood Disorders, Children’s Hospital Colorado, Aurora
| | - James A. Whitlock
- Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - Michael A. Pulsipher
- Transplantation and Cellular Therapy, Children's Hospital Los Angeles Cancer and Blood Diseases Institute, Los Angeles, 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
| | - Mignon L. Loh
- Department of Pediatrics, Benioff Children’s Hospital and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco
| |
Collapse
|
25
|
Karol SE, Yang JJ. Pharmacogenomics and ALL treatment: How to optimize therapy. Semin Hematol 2020; 57:130-136. [PMID: 33256902 DOI: 10.1053/j.seminhematol.2020.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 01/28/2023]
Abstract
Inherited genetic variations may alter drug sensitivity in patients with acute lymphoblastic leukemia, predisposing to adverse treatment side effects. In this review, we discuss evidence from children and young adults with acute lymphoblastic leukemia to review the available pharmacogenomic data with an emphasis on clinically actionable and emerging discoveries, for example, genetic variants in thiopurine methyltransferase and NUDT15 that alter 6-mercaptopurine dosing. We also highlight the need for ongoing pharmacogenomic research to validate the significance of recent findings. Further research in young adults, as well as with novel therapeutics, is needed to provide optimal therapy in future trials.
Collapse
Affiliation(s)
- Seth E Karol
- Departments of Oncology, St. Jude Children's Research Hospital, Memphis, TN.
| | - Jun J Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN
| |
Collapse
|
26
|
Jasinski S, De Los Reyes FA, Yametti GC, Pierro J, Raetz E, Carroll WL. Immunotherapy in Pediatric B-Cell Acute Lymphoblastic Leukemia: Advances and Ongoing Challenges. Paediatr Drugs 2020; 22:485-499. [PMID: 32860590 PMCID: PMC7537790 DOI: 10.1007/s40272-020-00413-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Leukemia, most commonly B-cell acute lymphoblastic leukemia (B-ALL), accounts for about 30% of childhood cancer diagnoses. While there have been dramatic improvements in childhood ALL outcomes, certain subgroups-particularly those who relapse-fare poorly. In addition, cure is associated with significant short- and long-term side effects. Given these challenges, there is great interest in novel, targeted approaches to therapy. A number of new immunotherapeutic agents have proven to be efficacious in relapsed or refractory disease and are now being investigated in frontline treatment regimens. Blinatumomab (a bispecific T-cell engager that targets cluster of differentiation [CD]-19) and inotuzumab ozogamicin (a humanized antibody-drug conjugate to CD22) have shown the most promise. Chimeric antigen receptor T (CAR-T) cells, a form of adoptive immunotherapy, rely on the transfer of genetically modified effector T cells that have the potential to persist in vivo for years, providing ongoing long-term disease control. In this article, we discuss the clinical biology and treatment of B-ALL with an emphasis on the role of immunotherapy in overcoming the challenges of conventional cytotoxic therapy. As immunotherapy continues to move into the frontline of pediatric B-ALL therapy, we also discuss strategies to address unique side effects associated with these agents and efforts to overcome mechanisms of resistance to immunotherapy.
Collapse
Affiliation(s)
- Sylwia Jasinski
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | | | - Gloria Contreras Yametti
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | - Joanna Pierro
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | - Elizabeth Raetz
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | - William L Carroll
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA.
| |
Collapse
|
27
|
Rubinstein JD, Krupski C, Nelson AS, O'Brien MM, Davies SM, Phillips CL. Chimeric Antigen Receptor T Cell Therapy in Patients with Multiply Relapsed or Refractory Extramedullary Leukemia. Biol Blood Marrow Transplant 2020; 26:e280-e285. [PMID: 32755637 DOI: 10.1016/j.bbmt.2020.07.036] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/30/2020] [Accepted: 07/30/2020] [Indexed: 12/29/2022]
Abstract
Autologous CD19-directed chimeric antigen receptor T lymphocyte (CAR-T) therapy is an approved and effective treatment for the management of patients with refractory and multiply relapsed B cell precursor acute lymphoblastic leukemia (B-ALL). Experience using this therapy in pediatric patients with extramedullary (EM) disease is limited, in part because these patients have frequently been excluded from clinical trials owing to concerns for an increased risk of immune effector cell-associated neurotoxicity syndrome (ICANS). We infused 7 patients with refractory or multiply relapsed B-ALL who presented with isolated EM relapse with tisagenlecleucel. Six patients had isolated central nervous system (CNS) leukemia, and 1 patient had an isolated testicular relapse. An initial complete response was seen in all patients, with 5 patients remaining in CAR-T-induced remission at a median of 18 months from first infusion. Reversible ICANS was seen in 1 patient with CNS leukemia. Durable B cell aplasia occurred in 3 patients, with a median time to B cell recovery of 6.5 months in the other patients. These data suggest that CAR-T therapy has promising safety and efficacy in treating EM leukemia, although definitive conclusions are limited by the small size of the cohort and limited follow-up period.
Collapse
Affiliation(s)
- Jeremy D Rubinstein
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| | - Christa Krupski
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Adam S Nelson
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Maureen M O'Brien
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Stella M Davies
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Christine L Phillips
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|