1
|
Kalwak K, Moser LM, Pötschger U, Bader P, Kleinschmidt K, Meisel R, Dalle JH, Yesilipek A, Balduzzi A, Krivan G, Goussetis E, Staciuk R, Sedlacek P, Pichler H, Svec P, Gabriel M, Güngör T, Bilic E, Buechner J, Renard M, Vettenranta K, Ifversen M, Diaz-de-Heredia C, Stein J, Toporski J, Bierings M, Peters C, Ansari M, Locatelli F. Comparable outcomes after busulfan- or treosulfan-based conditioning for allo-HSCT in children with ALL: results of FORUM. Blood Adv 2025; 9:741-751. [PMID: 39602342 PMCID: PMC11869852 DOI: 10.1182/bloodadvances.2024014548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/09/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
ABSTRACT The superiority of total body irradiation (TBI)-based vs chemotherapy conditioning for allogeneic hematopoietic stem cell transplantation (allo-HSCT) in children with acute lymphoblastic leukemia (ALL) has been established in the international, prospective phase-3 FORUM study, randomizing 417 patients aged 4-18 years in complete remission (CR), who received allo-HSCT from HLA-matched sibling or unrelated donors. Because of the unavailability of TBI in some regions and to accommodate individual contraindications, this study reports the prespecified comparison of outcomes of patients receiving busulfan (BU)- or treosulfan (TREO)-based regimens from 2013 to 2018. Overall, 180 and 128 patients received BU/thiotepa (THIO)/fludarabine (FLU) or TREO/THIO/FLU, respectively. Data were analyzed as of February 2023, with a median follow-up of 4.2 years (range, 0.3-9.1). 3-year overall survival was 0.71 (BU, 95% confidence interval [0.64-0.77]) and 0.72 (TREO, [0.63-0.79]) and 3-year event-free survival was 0.60 (BU, [0.53-0.67]) and 0.55 (TREO, [0.46-0.63]). The 3-year cumulative incidence of relapse (BU, 0.31 [0.25-0.38]; TREO, 0.36 [0.27-0.44]); and nonrelapse mortality (BU, 0.08 [0.05-0.13]; TREO, 0.09 [0.05-0.15]) were comparable. One case of fatal veno-occlusive disease occurred in each group. No significant differences in acute and chronic graft-versus-host disease (GVHD) or 3-year GVHD-free and relapse-free survival (BU, 0.48 [0.41-0.55]; TREO, 0.45 [0.37-0.54]) were recorded. Outcomes for patients in first and second CR were similar irrespective of the regimen. In conclusion, BU/THIO/FLU or TREO/THIO/FLU regimens can be an alternative to TBI for patients with ALL aged >4 years with contraindications or lack of access to TBI. This trial was registered at www.ClinicalTrials.gov as #NCT01949129.
Collapse
Affiliation(s)
- Krzysztof Kalwak
- Department of Pediatric Hematology, Oncology, and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - Laura M. Moser
- Division for Stem Cell Transplantation and Immunology, Department of Pediatrics, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | | | - Peter Bader
- Division for Stem Cell Transplantation and Immunology, Department of Pediatrics, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - Katharina Kleinschmidt
- Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation, University Children’s Hospital Regensburg, Regensburg, Germany
| | - Roland Meisel
- Division of Pediatric Stem Cell Therapy, Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Jean-Hugues Dalle
- Pediatric Hematology and Immunology Department, Robert Debré Hospital, Groupe Hospitalo-Universitaire Assistance Publique Hôpitaux de Paris Nord, Université Paris Cité, Paris, France
| | | | - Adriana Balduzzi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Pediatric Hematopoietic Stem Cell Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Gergely Krivan
- Pediatric Hematology and Stem Cell Transplantation Department, National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - Evgenios Goussetis
- Stem Cell Transplant Unit, Agia Sofia Children's Hospital, Athens, Greece
| | - Raquel Staciuk
- Hospital de Pediatría “Prof. Dr Juan P. Garrahan,” Buenos Aires, Argentina
| | - Petr Sedlacek
- Department of Pediatric Hematology and Oncology, University Hospital Motol, Prague, Czech Republic
| | - Herbert Pichler
- St. Anna Children’s Cancer Research Institute, Vienna, Austria
- Department of Pediatric Hematology and Oncology, St. Anna Children’s Hospital, Medical University of Vienna, Vienna, Austria
| | - Peter Svec
- Department of Pediatric Hematology and Oncology, National Institute of Children’s Diseases, Comenius University, Bratislava, Slovakia
| | | | - Tayfun Güngör
- Division of Hematology/Oncology/Immunology, Gene Therapy, and Stem Cell Transplantation, University Children's Hospital Zurich, Eleonore Foundation and Children’s Research Center, Zürich, Switzerland
| | - Ernest Bilic
- Division for Hematology and Oncology, Department of Pediatrics Zagreb, University Hospital Center, Zagreb, Croatia
| | - Jochen Buechner
- Department of Pediatric Hematology and Oncology, Oslo University Hospital, Oslo, Norway
| | - Marleen Renard
- Department of Paediatric Oncology, University Hospital Leuven, Leuven, Belgium
| | - Kim Vettenranta
- University of Helsinki and the Hospital for Children and Adolescents, University of Helsinki, Helsinki, Finland
| | - Marianne Ifversen
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Cristina Diaz-de-Heredia
- Division of Pediatric Hematology and Oncology, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Jerry Stein
- Schneider Children's Medical Center of Israel and Sackler Faculty of Medicine Tel Aviv University, Petah Tikva, Israel
| | - Jacek Toporski
- Department Cell Therapy and Allogeneic Stem Cell Transplant, Karolinska University Hospital, Stockholm, Sweden
| | - Marc Bierings
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Christina Peters
- St. Anna Children’s Cancer Research Institute, Vienna, Austria
- Department of Pediatric Hematology and Oncology, St. Anna Children’s Hospital, Medical University of Vienna, Vienna, Austria
| | - Marc Ansari
- CANSEARCH Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland
- Division of Pediatric Oncology and Hematology, Department of Women, Child and Adolescent, University Geneva Hospitals, Geneva, Switzerland
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
2
|
Ramdial J, Lin R, Thall PF, Valdez BC, Hosing C, Srour S, Popat U, Qazilbash M, Alousi A, Barnett M, Gulbis A, Shigle TL, Shpall EJ, Andersson BS, Nieto Y. High activity of the new myeloablative regimen of gemcitabine/clofarabine/busulfan for allogeneic transplant for aggressive lymphomas. Bone Marrow Transplant 2024; 59:1754-1762. [PMID: 39341929 PMCID: PMC11611727 DOI: 10.1038/s41409-024-02394-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/21/2024] [Accepted: 08/06/2024] [Indexed: 10/01/2024]
Abstract
Refractory aggressive lymphomas can be treated with allo-SCT, pursuing a graft-vs-lymphoma effect. While reduced intensity conditioning is safe, tumors often progress rapidly, indicating the need for more active conditioning regimens. The preclinical synergy we saw between gemcitabine (Gem), clofarabine (Clo) and busulfan (Bu) against lymphoma cell lines led us to study Gem/Clo/Bu clinically. Eligibility: age 12-65, refractory aggressive B-NHL, T-NHL or Hodgkin, with a matched donor. Infusional Gem was dose-escalated on days (d) -6 and -4 (475-975 mg/m2/day), followed by Clo (40 mg/m2/day) and Bu (target AUC, 4000 μMol min/day) (d -6 to -3). CD20+ tumors received rituximab. GVHD prophylaxis included ATG (MUD), tacrolimus and MMF. We compared their outcomes to matched-pair concurrent controls receiving Flu/Mel + matched allo-SCT. We enrolled 64 patients, median age 46 (17-63), 31 B-NHL/22 T-NHL/11 Hodgkin, 36 MSD/28 MUD (all PBPC), median 4 (2-10) prior therapies; 18 prior auto-SCT, 42 active diseases at allo-SCT (12 PD). Toxicities (mucositis and transaminitis) were manageable. Gem/Clo/Bu was myeloablative yielding early full donor chimerism. Grades II-IV/III-IV acute GVHD rates of 37% and 18%; chronic GVHD of 33% (13% severe); NRM at D100/1 year was 7% and 18%. ORR/CR rates: 78%/71% (B-NHL), 93%/93% (T-NHL), 67%/67% (Hodgkin). At a median follow-up of 60 (12-110) months, EFS/OS rates: 36%/47%. Gem/Clo/Bu patients had better median EFS (12 vs. 3 months, P = 0.001) and OS (25 vs. 7 months, P = 0.003) than 113 Flu/Mel matched-pair controls. The new myeloablative regimen Gem/Clo/Bu has limited toxicity and high activity in allo-SCT for aggressive lymphomas, yielding better outcomes than concurrent matched-pair controls receiving Flu/Mel.
Collapse
Affiliation(s)
- Jeremy Ramdial
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Ruitao Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Peter F Thall
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Benigno C Valdez
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chitra Hosing
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samer Srour
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Uday Popat
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Muzaffar Qazilbash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amin Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Melissa Barnett
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alison Gulbis
- Department of Pharmacy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Terri Lynn Shigle
- Department of Pharmacy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Borje S Andersson
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
3
|
Jansen SA, Cutilli A, de Koning C, van Hoesel M, Frederiks CL, Saiz Sierra L, Nierkens S, Mokry M, Nieuwenhuis EE, Hanash AM, Mocholi E, Coffer PJ, Lindemans CA. Chemotherapy-induced intestinal epithelial damage directly promotes galectin-9-driven modulation of T cell behavior. iScience 2024; 27:110072. [PMID: 38883813 PMCID: PMC11176658 DOI: 10.1016/j.isci.2024.110072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/05/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
The intestine is vulnerable to chemotherapy-induced damage due to the high rate of intestinal epithelial cell (IEC) proliferation. We have developed a human intestinal organoid-based 3D model system to study the direct effect of chemotherapy-induced IEC damage on T cell behavior. Exposure of intestinal organoids to busulfan, fludarabine, and clofarabine induced damage-related responses affecting both the capacity to regenerate and transcriptional reprogramming. In ex vivo co-culture assays, prior intestinal organoid damage resulted in increased T cell activation, proliferation, and migration. We identified galectin-9 (Gal-9) as a key molecule released by damaged organoids. The use of anti-Gal-9 blocking antibodies or CRISPR/Cas9-mediated Gal-9 knock-out prevented intestinal organoid damage-induced T cell proliferation, interferon-gamma release, and migration. Increased levels of Gal-9 were found early after HSCT chemotherapeutic conditioning in the plasma of patients who later developed acute GVHD. Taken together, chemotherapy-induced intestinal damage can influence T cell behavior in a Gal-9-dependent manner which may provide novel strategies for therapeutic intervention.
Collapse
Affiliation(s)
- Suze A. Jansen
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584CS, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
| | - Alessandro Cutilli
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
- Center of Molecular Medicine, University Medical Center Utrecht, Utrecht 3584CG, the Netherlands
| | - Coco de Koning
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584CS, the Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, 3584GX Utrecht, the Netherlands
| | - Marliek van Hoesel
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
| | - Cynthia L. Frederiks
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
- Center of Molecular Medicine, University Medical Center Utrecht, Utrecht 3584CG, the Netherlands
| | - Leire Saiz Sierra
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584CS, the Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, 3584GX Utrecht, the Netherlands
| | - Michal Mokry
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Department of Cardiology, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
| | - Edward E.S. Nieuwenhuis
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
- University College Roosevelt, Utrecht University, Middelburg 4331CB, the Netherlands
| | - Alan M. Hanash
- Departments of Medicine and Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY 10065, USA
| | - Enric Mocholi
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
- Center of Molecular Medicine, University Medical Center Utrecht, Utrecht 3584CG, the Netherlands
| | - Paul J. Coffer
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
- Center of Molecular Medicine, University Medical Center Utrecht, Utrecht 3584CG, the Netherlands
| | - Caroline A. Lindemans
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584CS, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
| |
Collapse
|
4
|
Gharial J, Guilcher G, Truong T, Shah R, Desai S, Rojas-Vasquez M, Kangarloo B, Lewis V. Busulfan with 400 centigray of total body irradiation and higher dose fludarabine: An alternative regimen for hematopoietic stem cell transplantation in pediatric acute lymphoblastic leukemia. Pediatr Blood Cancer 2024; 71:e30844. [PMID: 38217082 DOI: 10.1002/pbc.30844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 12/01/2023] [Accepted: 12/21/2023] [Indexed: 01/14/2024]
Abstract
BACKGROUND Hematopoietic stem cell transplantation can be curative for children with difficult-to-treat leukemia. The conditioning regimen utilized is known to influence outcomes. We report outcomes of the conditioning regimen used at the Alberta Children's Hospital, consisting of busulfan (with pharmacokinetic target of 3750 μmol*min/L/day ±10%) for 4 days, higher dose (250 mg/m2 ) fludarabine and 400 centigray (cGy) of total body irradiation. PROCEDURE This retrospective study involved children receiving transplant for acute lymphoblastic leukemia (ALL). It compared children who fell within the target range for busulfan with those who were either not measured or were measured and fell outside this range. All other treatment factors were identical. RESULTS Twenty-nine children (17 within target) were evaluated. All subjects engrafted neutrophils with a median [interquartile range] time of 14 days [8-30 days]. The cumulative incidence of acute graft-versus-host disease was 44.8% [95% confidence interval, CI: 35.6%-54.0%], while chronic graft-versus-host disease was noted in 16.0% [95% CI: 8.7%-23.3%]. At 2 years, the overall survival was 78.1% [95% CI: 70.8%-86.4%] and event-free survival was 74.7% [95% CI: 66.4%-83.0%]. Cumulative incidence of relapse was 11.3% [95% CI: 5.1%-17.5%]. There were no statistically significant differences in between the group that received targeted busulfan compared with the untargeted group. CONCLUSION Our conditioning regiment for children with ALL resulted in outcomes comparable to standard treatment with acceptable toxicities and significant reduction in radiation dose. Targeting busulfan dose in this cohort did not result in improved outcomes.
Collapse
Affiliation(s)
- Jaspreet Gharial
- Section of Pediatric Hematology Oncology and Bone Marrow Transplantation, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Gregory Guilcher
- Section of Pediatric Hematology Oncology and Bone Marrow Transplantation, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Tony Truong
- Section of Pediatric Hematology Oncology and Bone Marrow Transplantation, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Ravi Shah
- Section of Pediatric Hematology Oncology and Bone Marrow Transplantation, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Sunil Desai
- Division of Pediatric Hematology/Oncology & Palliative Care, Stollery Children's Hospital, Edmonton, Alberta, Canada
| | - Marta Rojas-Vasquez
- Division of Pediatric Hematology/Oncology & Palliative Care, Stollery Children's Hospital, Edmonton, Alberta, Canada
| | - Bill Kangarloo
- Pharmacokinetic Scientist, Alberta Blood and Marrow Transplant Program, Foothills Hospital, and Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | - Victor Lewis
- Section of Pediatric Hematology Oncology and Bone Marrow Transplantation, Alberta Children's Hospital, Calgary, Alberta, Canada
| |
Collapse
|
5
|
Zhao X, Xu Z, Li Z, Zhou X, Hu Y, Wang H. Intensified conditioning regimens with total marrow irradiation/etoposide/cyclophosphamide and busulfan/etoposide/cyclophosphamide overcome the impact of pre-transplant minimal residual disease on outcomes in high-risk acute lymphoblastic leukemia patients in complete remission. Cancer Med 2024; 13:e6897. [PMID: 38164654 PMCID: PMC10807553 DOI: 10.1002/cam4.6897] [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: 08/17/2023] [Revised: 12/12/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024] Open
Abstract
PURPOSE Among high-risk acute lymphoblastic leukemia (ALL) patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT), those with positive minimal residual disease (MRD) are susceptible to poor outcomes. Therefore, it is necessary to determine the most suitable preparatory regimen for these patients. METHODS Data were analyzed from 141 patients who received allo-HSCT and were diagnosed with high-risk ALL. These patients underwent intensified conditioning regimens, including either total marrow and lymphoid irradiation (TMLI)-etoposide (VP16)-cyclophosphamide (CY) or busulfan (BU)-VP16-CY between October 2016 and November 2022. A total of 141 individuals were in complete remission (CR) before transplantation and, among all patients, 90 individuals exhibited a negative MRD status and 51 patients had a positive MRD status. RESULTS In patients who tested negative for MRD, the incidence of relapse within a 2-year timeframe was 25.0% (24.8%-25.5%), compared with 32.2% (31.2%-33.2%) in MRD-positive patients; however, this difference was not statistically significant. There were no significant differences in the 2-year disease-free survival (DFS) and 2-year overall survival (OS) rates between the MRD-negative and MRD-positive groups (DFS: 67.2% (57.9%-78.1%) vs. 55.5% (42.6%-72.3%); OS: 69.0% (61.9%-88.2%) vs. 66.7% (53.9%-82.5%)). Furthermore, no notable variations were observed in the occurrence of transplant-related mortality (TRM) and graft-versus-host disease (GVHD) across the two groups. CONCLUSION This study reveals the benefits of TMLI-VP16-CY and BU-VP16-CY conditioning regimens in high-risk ALL patients with CR and MRD-positive status. A large-scale prospective clinical trial is warranted in the future.
Collapse
Affiliation(s)
- Xiaoyan Zhao
- Department of Hematology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ziwei Xu
- Department of Hematology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ziying Li
- Department of Pediatrics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xi Zhou
- Department of Pathology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yu Hu
- Department of Hematology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Huafang Wang
- Department of Hematology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
6
|
Jansen SA, Cutilli A, de Koning C, van Hoesel M, Sierra LS, Nierkens S, Mokry M, Nieuwenhuis EES, Hanash AM, Mocholi E, Coffer PJ, Lindemans CA. Chemotherapy-induced intestinal injury promotes Galectin-9-driven modulation of T cell function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.30.538862. [PMID: 37163028 PMCID: PMC10168344 DOI: 10.1101/2023.04.30.538862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The intestine is vulnerable to chemotherapy-induced toxicity due to its high epithelial proliferative rate, making gut toxicity an off-target effect in several cancer treatments, including conditioning regimens for allogeneic hematopoietic cell transplantation (allo-HCT). In allo-HCT, intestinal damage is an important factor in the development of Graft-versus-Host Disease (GVHD), an immune complication in which donor immune cells attack the recipient's tissues. Here, we developed a novel human intestinal organoid-based 3D model system to study the direct effect of chemotherapy-induced intestinal epithelial damage on T cell behavior. Chemotherapy treatment using busulfan, fludarabine, and clofarabine led to damage responses in organoids resulting in increased T cell migration, activation, and proliferation in ex- vivo co-culture assays. We identified galectin-9 (Gal-9), a beta-galactoside-binding lectin released by damaged organoids, as a key molecule mediating T cell responses to damage. Increased levels of Gal-9 were also found in the plasma of allo-HCT patients who later developed acute GVHD, supporting the predictive value of the model system in the clinical setting. This study highlights the potential contribution of chemotherapy-induced epithelial damage to the pathogenesis of intestinal GVHD through direct effects on T cell activation and trafficking promoted by galectin-9.
Collapse
|
7
|
Connor MP, Loren AW, Hexner EO, Martin ME, Gill SI, Luger SM, Mangan JK, Perl AE, McCurdy SR, Pratz KW, Timlin C, Freyer CW, Carulli A, Catania C, Smith J, Hollander L, Zebrowski AM, Stadtmauer EA, Porter DL, Frey NV. Clofarabine and Busulfan Myeloablative Conditioning in Allogeneic Hematopoietic Cell Transplantation for Patients With Active Myeloid Malignancies. Transplant Cell Ther 2023; 29:113-118. [PMID: 36336258 DOI: 10.1016/j.jtct.2022.10.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/19/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
Abstract
Patients with refractory or relapsed and refractory myeloid malignancies have a poor prognosis. Allogeneic hematopoietic cell transplantation (HCT) with myeloablative conditioning (MAC) in patients with active, chemotherapy-refractory myeloid disease is historically associated with high rates of relapse and nonrelapse mortality (NRM). A MAC regimen combining clofarabine with busulfan (Clo/Bu4) has been reported to exhibit antileukemic activity with acceptable toxicity in patients age ≤70 years. Here we describe the clinical outcomes of a real-world population of patients with active myeloid malignancies undergoing allogeneic HCT with Clo/Bu4 MAC. In a single-center retrospective descriptive analysis, we identified patients who underwent HCT for myeloid malignancies not in remission using Clo/Bu4 MAC between 2012 and 2020. We report event-free survival (EFS) and overall survival (OS), cumulative incidences of relapse and NRM, and the incidence and severity of acute and chronic graft-versus-host disease (GVHD). We identified 69 patients with a median age of 60 years (range, 22 to 70 years). Most patients had relapsed/refractory or primary refractory acute myelogenous leukemia (AML; n = 55) or refractory myelodysplastic syndrome (MDS; n = 12); 1 patient had chronic myelogenous leukemia, and 1 patient had a blastic plasmacytoid dendritic cell neoplasm. Fifty patients (72.5%) had complete remission at day 100 post-transplantation. Two-year EFS and OS were 30% (95% confidence interval [CI], 20% to 44%) and 40% (95% CI, 29% to 54%), respectively. Patients with AML had a 2-year EFS and OS of 28% (95% CI, 18% to 44%) and 38% (95% CI, 27% to 54%), respectively; those with MDS had a 2-year EFS and OS of 47% (95% CI, 25% to 88%) and 56% (95% CI, 33% to 94%), respectively. The cumulative incidence of relapse at 2 years was 39% (95% CI, 27% to 51%) for all patients, including 45% (95% CI, 31% to 58%) in the patients with AML and 18% (95% CI, 2% to 45%) in those with MDS. NRM at 2 years was 31% (95% CI, 20% to 42%), including 27% (95% CI, 15% to 39%) in patients with AML and 35% (95% CI, 10% to 63%) in those with MDS. The total incidence of acute GVHD (aGVHD) of any severity was 80%, and the incidence of grade III-IV aGVHD was 22%. In patients who achieved remission, those who required systemic immunosuppression for aGVHD (58%) had poorer 2-year EFS (29% versus 54%; P = .05) and 2-year OS (39% versus 70%; P = .04) compared to those who did not. The 2-year cumulative incidence of chronic GVHD was 44% (95% CI, 28% to 58%). Clo/Bu4 MAC followed by allogeneic HCT for patients with active myeloid malignancies is an effective transplantation strategy for patients up to age 70, particularly those with advanced MDS. The high incidence of and poor outcomes associated with aGVHD highlight the importance of optimizing preventative strategies.
Collapse
Affiliation(s)
- Matthew P Connor
- Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Alison W Loren
- Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth O Hexner
- Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mary Ellen Martin
- Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Saar I Gill
- Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Selina M Luger
- Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - James K Mangan
- Moores Cancer Center at the University of California, San Diego, California
| | - Alexander E Perl
- Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shannon R McCurdy
- Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Keith W Pratz
- Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Colleen Timlin
- Department of Pharmacy, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Craig W Freyer
- Department of Pharmacy, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alison Carulli
- Department of Pharmacy, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christopher Catania
- Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jacqueline Smith
- Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lauren Hollander
- Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alexis M Zebrowski
- Department of Emergency Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Edward A Stadtmauer
- Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - David L Porter
- Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Noelle V Frey
- Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
8
|
Logan AC. SOHO State of the Art Updates and Next Questions: Novel Transplant and Post-Transplant Options in Acute Lymphoblastic Leukemia. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:569-574. [PMID: 35410757 DOI: 10.1016/j.clml.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 06/14/2023]
Abstract
Allogeneic hematopoietic cell transplantation (alloHCT) is a potentially curative treatment approach for patients with high-risk acute lymphoblastic leukemia (ALL). Despite development of several novel therapies targeting B-cell ALL, alloHCT continues to play an essential role in management, but the identification of patients who are most likely to benefit from alloHCT in first or subsequent remissions continues to evolve. Broader donor options, including haploidentical donors and umbilical cord blood, have enabled alloHCT for more patients, but improvements in front-line therapy and increasing use of high-sensitivity measurable residual disease (MRD) quantification continue to modify the calculus for selecting which patients require transplantation. MRD quantification has become increasingly important as a prognostic indicator, as well as a trigger for therapeutic intervention, since the achievement of MRD negative complete remission is well-established to be associated with improved transplant outcomes. ALL remains the only malignancy with approved therapy for MRD positivity after achievement of remission, and use of Blinatumomab in this setting currently appears to be most effective when used as a bridge-to-transplant, rather than a destination or purely consolidative therapy. Expanding options for those with relapsed/refractory disease, including chimeric antigen receptor (CAR)-T cells, also render more patient in suitably deep remissions to enable alloHCT with a high likelihood of success. It remains unclear whether CAR-T cell therapies may obviate the need for alloHCT in some patients, and currently available data suggest there remains a role for alloHCT after CAR-T. Together, these therapeutic advances appear to be improving post-transplant outcomes. Nevertheless, more remains to be studied regarding how to optimize use of available and emerging cellular and immune modulating therapies to maximize the likelihood of long-term post-alloHCT remission in high-risk ALL.
Collapse
Affiliation(s)
- Aaron C Logan
- University of California, San Francisco, Division of Hematology, Blood and Marrow Transplantation, and Cellular Therapy, San Francisco, CA.
| |
Collapse
|
9
|
Alatrash G, Saberian C, Bassett R, Thall PF, Ledesma C, Lu Y, Daher M, Valdez BC, Kawedia J, Popat U, Mehta R, Oran B, Nieto Y, Olson A, Anderlini P, Marin D, Hosing C, Alousi AM, Shpall EJ, Rondon G, Chen J, Qazilbash M, Champlin RE, Andersson BS, Kebriaei P. Vorinostat combined with Busulfan, Fludarabine, and Clofarabine Conditioning Regimen for Allogeneic Hematopoietic Stem Cell Transplantation in Patients with Acute Leukemia: Long-term Study Outcomes. Transplant Cell Ther 2022; 28:501.e1-501.e7. [DOI: 10.1016/j.jtct.2022.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/15/2022] [Accepted: 05/14/2022] [Indexed: 11/30/2022]
|
10
|
Ben Hassine K, Powys M, Svec P, Pozdechova M, Versluys B, Ansari M, Shaw PJ. Total Body Irradiation Forever? Optimising Chemotherapeutic Options for Irradiation-Free Conditioning for Paediatric Acute Lymphoblastic Leukaemia. Front Pediatr 2021; 9:775485. [PMID: 34956984 PMCID: PMC8705537 DOI: 10.3389/fped.2021.775485] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/21/2021] [Indexed: 12/15/2022] Open
Abstract
Total-body irradiation (TBI) based conditioning prior to allogeneic hematopoietic stem cell transplantation (HSCT) is generally regarded as the gold-standard for children >4 years of age with acute lymphoblastic leukaemia (ALL). Retrospective studies in the 1990's suggested better survival with irradiation, confirmed in a small randomised, prospective study in the early 2000's. Most recently, this was reconfirmed by the early results of the large, randomised, international, phase III FORUM study published in 2020. But we know survivors will suffer a multitude of long-term sequelae after TBI, including second malignancies, neurocognitive, endocrine and cardiometabolic effects. The drive to avoid TBI directs us to continue optimising irradiation-free, myeloablative conditioning. In chemotherapy-based conditioning, the dominant myeloablative effect is provided by the alkylating agents, most commonly busulfan or treosulfan. Busulfan with cyclophosphamide is a long-established alternative to TBI-based conditioning in ALL patients. Substituting fludarabine for cyclophosphamide reduces toxicity, but may not be as effective, prompting the addition of a third agent, such as thiotepa, melphalan, and now clofarabine. For busulfan, it's wide pharmacokinetic (PK) variability and narrow therapeutic window is well-known, with widespread use of therapeutic drug monitoring (TDM) to individualise dosing and control the cumulative busulfan exposure. The development of first-dose selection algorithms has helped achieve early, accurate busulfan levels within the targeted therapeutic window. In the future, predictive genetic variants, associated with differing busulfan exposures and toxicities, could be employed to further tailor individualised busulfan-based conditioning for ALL patients. Treosulfan-based conditioning leads to comparable outcomes to busulfan-based conditioning in paediatric ALL, without the need for TDM to date. Future PK evaluation and modelling may optimise therapy and improve outcome. More recently, the addition of clofarabine to busulfan/fludarabine has shown encouraging results when compared to TBI-based regimens. The combination shows activity in ALL as well as AML and deserves further evaluation. Like busulfan, optimization of chemotherapy conditioning may be enhanced by understanding not just the PK of clofarabine, fludarabine, treosulfan and other agents, but also the pharmacodynamics and pharmacogenetics, ideally in the context of a single disease such as ALL.
Collapse
Affiliation(s)
- Khalil Ben Hassine
- Cansearch Research Platform for Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Madeleine Powys
- Blood Transplant and Cell Therapies, Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Peter Svec
- Department of Pediatric Hematology and Oncology, Comenius University, Bratislava, Slovakia.,Bone Marrow Transplantation Unit, National Institute of Children's Diseases, Bratislava, Slovakia
| | - Miroslava Pozdechova
- Department of Pediatric Hematology and Oncology, Comenius University, Bratislava, Slovakia.,Bone Marrow Transplantation Unit, National Institute of Children's Diseases, Bratislava, Slovakia
| | | | - Marc Ansari
- Cansearch Research Platform for Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Division of Pediatric Oncology and Hematology, Department of Women, Child and Adolescent, University Geneva Hospitals, Geneva, Switzerland
| | - Peter J Shaw
- Blood Transplant and Cell Therapies, Children's Hospital at Westmead, Sydney, NSW, Australia.,Speciality of Child and Adolescent Health, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
11
|
Peters C, Dalle JH, Locatelli F, Poetschger U, Sedlacek P, Buechner J, Shaw PJ, Staciuk R, Ifversen M, Pichler H, Vettenranta K, Svec P, Aleinikova O, Stein J, Güngör T, Toporski J, Truong TH, Diaz-de-Heredia C, Bierings M, Ariffin H, Essa M, Burkhardt B, Schultz K, Meisel R, Lankester A, Ansari M, Schrappe M, von Stackelberg A, Balduzzi A, Corbacioglu S, Bader P. Total Body Irradiation or Chemotherapy Conditioning in Childhood ALL: A Multinational, Randomized, Noninferiority Phase III Study. J Clin Oncol 2020; 39:295-307. [PMID: 33332189 PMCID: PMC8078415 DOI: 10.1200/jco.20.02529] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Total body irradiation (TBI) before allogeneic hematopoietic stem cell transplantation (HSCT) in pediatric patients with acute lymphoblastic leukemia (ALL) is efficacious, but long-term side effects are concerning. We investigated whether preparative combination chemotherapy could replace TBI in such patients.
Collapse
Affiliation(s)
- Christina Peters
- St. Anna Children's Hospital, Children's Cancer Research Institute, University Vienna, Vienna, Austria
| | - Jean-Hugues Dalle
- Hôpital Robert Debré, GH APHP-Nord Université de Paris, Paris, France
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza University of Rome, Rome, Italy
| | | | - Petr Sedlacek
- Department of Pediatric Hematology and Oncology, Motol University Hospital, Prague, Czech Republic
| | - Jochen Buechner
- Department of Pediatric Hematology and Oncology, Oslo University Hospital, Oslo, Norway
| | - Peter J Shaw
- The Children`s Hospital at Westmead, Sydney, Australia
| | | | | | - Herbert Pichler
- St. Anna Children's Hospital, Children's Cancer Research Institute, University Vienna, Vienna, Austria
| | - Kim Vettenranta
- Children's Hospital, University of Helsinki, Helsinki, Finland
| | - Peter Svec
- National Institute of Children's Diseases, Bratislava, Slovakia
| | - Olga Aleinikova
- Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Borovlyani, Belarus
| | - Jerry Stein
- Schneider Children's Medical Center of Israel, Sackler Faculty of Medicine, Tel Aviv University, Petach-Tikva, Israel
| | | | | | - Tony H Truong
- Alberta Children's Hospital Calgary, Calgary, Alberta, Canada
| | | | - Marc Bierings
- Princess Máxima Center for Pediatric Oncology, Bilthoven, the Netherlands
| | | | - Mohammed Essa
- King Abdullah Specialist Children's Hospital, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | | | - Kirk Schultz
- University of British Columbia, Vancouver, British Columbia, Canada
| | - Roland Meisel
- Division of Pediatric Stem Cell Therapy, Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Arjan Lankester
- Willem-Alexander Children's Hospital, Leiden, the Netherlands
| | - Marc Ansari
- Geneva University Hospital, Geneva, Switzerland
| | | | | | | | | | | | | | | | | | - Peter Bader
- Goethe University, University Hospital Frankfurt, Department for Children and Adolescents, Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Frankfurt am Main, Germany
| |
Collapse
|
12
|
Friend BD, Bailey-Olson M, Melton A, Shimano KA, Kharbanda S, Higham C, Winestone LE, Huang J, Stieglitz E, Dvorak CC. The impact of total body irradiation-based regimens on outcomes in children and young adults with acute lymphoblastic leukemia undergoing allogeneic hematopoietic stem cell transplantation. Pediatr Blood Cancer 2020; 67:e28079. [PMID: 31724815 DOI: 10.1002/pbc.28079] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/08/2019] [Accepted: 10/27/2019] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Total body irradiation (TBI)-based conditioning is the standard of care in the treatment of acute lymphoblastic leukemia (ALL) that requires allogeneic hematopoietic stem cell transplantation (HSCT). However, TBI is known to be associated with an increased risk of late effects, and therefore, non-TBI regimens have also been utilized successfully. A recent study showed that patients that were next-generation sequencing-minimal residual disease (NGS-MRD) negative prior to allogeneic HSCT had a very low risk of relapse, and perhaps could avoid exposure to TBI without compromising disease control. We examined outcomes at our institution in patients that received a TBI or non-TBI regimen, as well as explored the impact of NGS-MRD status in predicting risk of relapse post transplant. PROCEDURES This retrospective analysis included 57 children and young adults with ALL that received their first myeloablative allogeneic HSCT from 2012 to 2017 at the University of California San Francisco. Our primary endpoint was the cumulative incidence of relapse at 3 years post transplant. RESULTS We demonstrated similar cumulative incidence of relapse for patients treated with either a TBI or non-TBI conditioning regimen, while NGS-MRD positivity prior to transplant was highly predictive of relapse. The presence of acute graft-versus-host disease was associated with decreased relapse rates, particularly among patients that received a TBI conditioning regimen and patients that were NGS-MRD positive prior to HSCT. CONCLUSIONS Our data suggest that the decision to use either a TBI or non-TBI regimens in ALL should depend on NGS-MRD status, with conditioning regimens based on TBI reserved for patients that cannot achieve NGS-MRD negativity prior to allogeneic HSCT.
Collapse
Affiliation(s)
- Brian D Friend
- Division of Pediatric Allergy, Immunology, and Blood & Marrow Transplantation, UCSF Benioff Children's Hospital, San Francisco, California.,Department of Pediatrics, Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas
| | - Mara Bailey-Olson
- Division of Pediatric Allergy, Immunology, and Blood & Marrow Transplantation, UCSF Benioff Children's Hospital, San Francisco, California
| | - Alexis Melton
- Division of Pediatric Allergy, Immunology, and Blood & Marrow Transplantation, UCSF Benioff Children's Hospital, San Francisco, California
| | - Kristin A Shimano
- Division of Pediatric Allergy, Immunology, and Blood & Marrow Transplantation, UCSF Benioff Children's Hospital, San Francisco, California.,Division of Pediatric Hematology/Oncology, UCSF Benioff Children's Hospital, San Francisco, California
| | - Sandhya Kharbanda
- Division of Pediatric Allergy, Immunology, and Blood & Marrow Transplantation, UCSF Benioff Children's Hospital, San Francisco, California
| | - Christine Higham
- Division of Pediatric Allergy, Immunology, and Blood & Marrow Transplantation, UCSF Benioff Children's Hospital, San Francisco, California
| | - Lena E Winestone
- Division of Pediatric Allergy, Immunology, and Blood & Marrow Transplantation, UCSF Benioff Children's Hospital, San Francisco, California
| | - James Huang
- Division of Pediatric Allergy, Immunology, and Blood & Marrow Transplantation, UCSF Benioff Children's Hospital, San Francisco, California.,Division of Pediatric Hematology/Oncology, UCSF Benioff Children's Hospital, San Francisco, California
| | - Elliot Stieglitz
- Division of Pediatric Hematology/Oncology, UCSF Benioff Children's Hospital, San Francisco, California
| | - Christopher C Dvorak
- Division of Pediatric Allergy, Immunology, and Blood & Marrow Transplantation, UCSF Benioff Children's Hospital, San Francisco, California
| |
Collapse
|
13
|
Shah S, Martin A, Turner M, Cong Z, Zaman F, Stein A. A systematic review of outcomes after stem cell transplantation in acute lymphoblastic leukemia with or without measurable residual disease. Leuk Lymphoma 2020; 61:1052-1062. [DOI: 10.1080/10428194.2019.1709834] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
| | - Amber Martin
- EVIDERA, Evidence, Synthesis, Modeling, and Communications, Waltham, MA, USA
| | - Monica Turner
- EVIDERA, Evidence, Synthesis, Modeling, and Communications, Waltham, MA, USA
| | - Ze Cong
- Amgen Inc., Thousand Oaks, CA, USA
| | | | - Anthony Stein
- City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
14
|
Krakow EF, Gyurkocza B, Storer BE, Chauncey TR, McCune JS, Radich JP, Bouvier ME, Estey EH, Storb R, Maloney DG, Sandmaier BM. Phase I/II multisite trial of optimally dosed clofarabine and low-dose TBI for hematopoietic cell transplantation in acute myeloid leukemia. Am J Hematol 2020; 95:48-56. [PMID: 31637757 DOI: 10.1002/ajh.25665] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 11/11/2022]
Abstract
Clofarabine is an immunosuppressive purine nucleoside analog that may have better anti-leukemic activity than fludarabine. We performed a prospective phase I/II multisite trial of clofarabine with 2 Gy total body irradiation as non-myeloablative conditioning for allogeneic hematopoietic cell transplantation in adults with acute myeloid leukemia who were unfit for more intense regimens. Our main objective was to improve the 6-month relapse rate following non-myeloablative conditioning, while maintaining historic rates of non-relapse mortality (NRM) and engraftment. Forty-four patients, 53 to 74 (median: 69) years, were treated with clofarabine at 150 to 250 mg/m2 , of whom 36 were treated at the maximum protocol-specified dose. One patient developed multifactorial acute kidney injury and another developed multiorgan failure, but no other grade 3 to 5 non-hematologic toxicities were observed. All patients fully engrafted. The 6-month relapse rate was 16% (95% CI, 5%-27%) among all patients and 14% (95% CI, 3%-26%) among high-risk patients treated at the maximum dose, meeting the pre-specified primary efficacy endpoint. Overall survival was 55% (95% CI, 40%-70%) and leukemia-free survival was 52% (95% CI, 37%-67%) at 2 years. Compared to a historical high-risk cohort treated with the combination of fludarabine at 90 mg/m2 and 2 Gy TBI, protocol patients treated with the clofarabine-TBI regimen had lower rates of overall mortality (HR of 0.50, 95% CI, 0.28-0.91), disease progression or death (HR 0.48, 95% CI, 0.27-0.85), and morphologic relapse (HR 0.30, 95% CI, 0.13-0.69), and comparable NRM (HR 0.85, 95% CI 0.36-2.00). The combination of clofarabine with TBI warrants further investigation in patients with high-risk AML.
Collapse
Affiliation(s)
- Elizabeth F. Krakow
- Clinical Research DivisionFred Hutchinson Cancer Research Center Seattle Washington
- Department of MedicineUniversity of Washington Seattle Washington
| | - Boglarka Gyurkocza
- Clinical Research DivisionFred Hutchinson Cancer Research Center Seattle Washington
| | - Barry E. Storer
- Clinical Research DivisionFred Hutchinson Cancer Research Center Seattle Washington
| | - Thomas R. Chauncey
- Clinical Research DivisionFred Hutchinson Cancer Research Center Seattle Washington
- Department of MedicineUniversity of Washington Seattle Washington
- Bone Marrow Transplant Unit, VA Puget Sound Health Care System Seattle Washington
| | - Jeannine S. McCune
- Clinical Research DivisionFred Hutchinson Cancer Research Center Seattle Washington
- Department of PharmaceuticsUniversity of Washington Seattle Washington
| | - Jerald P. Radich
- Clinical Research DivisionFred Hutchinson Cancer Research Center Seattle Washington
- Department of MedicineUniversity of Washington Seattle Washington
| | - Michelle E. Bouvier
- Clinical Research DivisionFred Hutchinson Cancer Research Center Seattle Washington
| | - Elihu H. Estey
- Clinical Research DivisionFred Hutchinson Cancer Research Center Seattle Washington
- Department of MedicineUniversity of Washington Seattle Washington
| | - Rainer Storb
- Clinical Research DivisionFred Hutchinson Cancer Research Center Seattle Washington
- Department of MedicineUniversity of Washington Seattle Washington
| | - David G Maloney
- Clinical Research DivisionFred Hutchinson Cancer Research Center Seattle Washington
- Department of MedicineUniversity of Washington Seattle Washington
| | - Brenda M. Sandmaier
- Clinical Research DivisionFred Hutchinson Cancer Research Center Seattle Washington
- Department of MedicineUniversity of Washington Seattle Washington
| |
Collapse
|
15
|
Peccatori J, Mastaglio S, Giglio F, Greco R, Crocchiolo R, Patriarca F, Forno B, Deola S, Assanelli A, Lupo Stanghellini MT, Marcatti M, Zecca M, Cortelazzo S, Fanin R, Fagioli F, Locatelli F, Ciceri F. Clofarabine and Treosulfan as Conditioning for Matched Related and Unrelated Hematopoietic Stem Cell Transplantation: Results from the Clo3o Phase II Trial. Biol Blood Marrow Transplant 2019; 26:316-322. [PMID: 31605823 DOI: 10.1016/j.bbmt.2019.09.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/17/2019] [Accepted: 09/30/2019] [Indexed: 10/25/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) can be curative for patients with hematologic malignancies. The ideal conditioning regimen before allo-HSCT has not been established. We conducted a Phase II study to evaluate the tolerability and efficacy of clofarabine and treosulfan as conditioning regimen before allo-HSCT. The primary objective was to evaluate the cumulative incidence of nonrelapse mortality (NRM) on day +100. Forty-four patients (36 with acute myelogenous leukemia, 5 with acute lymphoblastic leukemia, 3 with myelodysplastic syndromes) were enrolled. The median patient age was 47 years, and the median duration of follow-up was 27 months. The conditioning regimen was based on clofarabine 40 mg/m2 (days -6 to -2) and treosulfan 14 g/m2 (days -6 to -4). Allogeneic hematopoietic stem cells were derived from a sibling (n = 22) or a well-matched unrelated donor (n = 22). Graft-versus-host disease (GVHD) prophylaxis consisted of antithymocyte globulin, rituximab, cyclosporine, and a short-course of methotrexate. The regimen allowed for rapid engraftment and a 100-day NRM of 18%, due mainly to bacterial infections. The incidences of grade II-IV acute GVHD and chronic GVHD were 16% and 19%, respectively. The rates of overall survival (OS), progression-free survival, and relapse at 2 years were 51%, 31%, and 50%, respectively. Significantly different outcomes were observed between patients with low-intermediate and patients with high-very high Disease Risk Index (DRI) scores (1-year OS, 78% and 24%, respectively). Our findings show that the use of treosulfan and clofarabine as a conditioning regimen for allo-HSCT is feasible, with a 78% 1-year OS in patients with a low-intermediate DRI score. However, 1-year NRM was 18%, and despite the intensified conditioning regimen, relapse incidence remains a major issue in patients with poor prognostic risk factors.
Collapse
Affiliation(s)
- Jacopo Peccatori
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sara Mastaglio
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Giglio
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Raffaella Greco
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Francesca Patriarca
- Carlo Melzi Hematology and Cellular Therapy Unit, Azienda Sanitaria Universitaria Integrata di Udine, Undine, Italy
| | - Barbara Forno
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sara Deola
- Department of Hematology, Ospedale Regionale, Bolzano, Italy
| | - Andrea Assanelli
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Magda Marcatti
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Zecca
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Renato Fanin
- Carlo Melzi Hematology and Cellular Therapy Unit, Azienda Sanitaria Universitaria Integrata di Udine, Undine, Italy
| | - Franca Fagioli
- Pediatric Onco-Hematology, University of Torino, Torino, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Fabio Ciceri
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
16
|
Dermatologic Conditions of the Early Post-Transplant Period in Hematopoietic Stem Cell Transplant Recipients. Am J Clin Dermatol 2019; 20:55-73. [PMID: 30298481 DOI: 10.1007/s40257-018-0391-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hematopoietic stem cell transplants (HSCTs) are used to treat a variety of conditions, including hematologic malignancies, bone marrow failure syndromes, and immunodeficiencies. Over 60,000 HSCTs are performed annually worldwide, and the numbers continue to increase. Indeed, as new conditioning regimens develop, more and more individuals, including those of older age, will be eligible for transplants. Nevertheless, although HSCTs are clearly a life-saving and necessary treatment for thousands of patients per year, there is still substantial morbidity and mortality associated with the procedure. Of note, skin eruptions in the post-HSCT period are frequent and often significantly reduce quality of life in recipients. Moreover, these cutaneous findings sometimes herald an underlying systemic condition, presenting possible opportunities for timelier intervention. Dermatologists therefore play a vital role in distinguishing life-threatening conditions from benign issues and prompting recognition of critical complications earlier in their course. This article aims to review the major dermatologic conditions occurring in the early post-HSCT period.
Collapse
|
17
|
Le Bourgeois A, Labopin M, Leclerc M, de Latour RP, Bourhis JH, Ceballos P, Orvain C, Wallet HL, Bilger K, Blaise D, Rubio MT, Guillaume T, Mohty M, Chevallier P. Clofarabine/busulfan-based reduced intensity conditioning regimens provides very good survivals in acute myeloid leukemia patients in complete remission at transplant: a retrospective study on behalf of the SFGM-TC. Oncotarget 2018; 9:36603-36612. [PMID: 30564300 PMCID: PMC6290956 DOI: 10.18632/oncotarget.26391] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/01/2018] [Indexed: 11/25/2022] Open
Abstract
Background Clofarabine has been proved to have higher anti-leukemic myeloid activity compared to fludarabine, a drug extensively used as part of reduced intensity conditioning (RIC) for allogeneic stem cell transplantation (allo-SCT). Results Eighty-four patients were included. The majority of patients had acute myeloid leukemia (AML, n = 63). Sixty-one patients were in complete remission (AML n = 55). With a median follow up of 31 months (range: 5.7-74.1), 2-year overall (OS) and disease-free (DFS) survivals, relapse incidence (RI), non-relapse mortality (NRM) and graft-versus-host disease (GVHD)/relapse free survival (GRFS) were 64.5% (53.8-75.2); 57.2% (46.2-68.2); 27.7% (18.2-37.9); 15.1% (8.2-23.9) and 43.6% (32.5-54.7), respectively. Considering AML in remission, 2-year OS, DFS, RI, NRM and GRFS were 74.2% (62-86.5); 66.8% (53.6-79.9); 23.4% (12.7-36); 9.8% (3.5-19.9) and 50.9% (36.9-64.9), respectively. Two-year outcomes were similar between CloB2A1 and CloB2A2 sub-groups. In multivariate analysis, active disease at transplant was the only factor adversely impacting 2 years outcomes. Conclusions CloB2A2/A1 RIC regimen provides very good results for AML patients allografted in CR and could be retained as a new RIC platform for these patients. Materials and Methods This was a retrospective study including all patients who received a clofarabine/busulfan based RIC allo-SCT for myeloid malignancies and reported within the SFGM-TC registry. RIC regimen consisted of clofarabine 30 mg/m2/day 4 to 5 days (Clo), busulfan 3.2 mg/kg/day 2 days (B2) and 2.5 mg/kg/day of rabbit anti-thymocyte globulin 1 or 2 days (A1 or A2). The primary objective of the study was to report the main outcomes of the whole cohort at 2 years.
Collapse
Affiliation(s)
| | - Myriam Labopin
- Department of Hematology, Hôpital Saint Antoine, Paris, France
| | - Mathieu Leclerc
- Department of Hematology, Hôpital Henri Mondor, Créteil, France
| | | | | | - Patrice Ceballos
- Department of Hematology, CHU de Montpellier, Montpellier, France
| | | | | | - Karin Bilger
- Department of Hematology, CHU Strasbourg, Strasbourg, France
| | - Didier Blaise
- Department of Hematology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli Calmettes, Marseille, France
| | | | | | - Mohamad Mohty
- Department of Hematology, Hôpital Saint Antoine, Paris, France
| | | | - on behalf of Société Francophone de Greffe de Moelle et de Thérapie Cellulaire
- Department of Hematology, CHU Hôtel Dieu, Nantes, France
- Department of Hematology, Hôpital Saint Antoine, Paris, France
- Department of Hematology, Hôpital Henri Mondor, Créteil, France
- Department of Hematology, Hôpital Saint Louis, Université Paris 7, Denis Diderot, Paris, France
- Department of Hematology, Hôpital Gustave Roussy, Paris, France
- Department of Hematology, CHU de Montpellier, Montpellier, France
- Department of Hematology, CHU d’Angers, Angers, France
- Department of Hematology, Centre Hospitalier Lyon Sud, Lyon, France
- Department of Hematology, CHU Strasbourg, Strasbourg, France
- Department of Hematology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli Calmettes, Marseille, France
- Department of Hematology, CHU Nancy, Nancy, France
| |
Collapse
|
18
|
Valdez BC, Tang X, Li Y, Murray D, Liu Y, Popat U, Champlin RE, Andersson BS. Epigenetic modification enhances the cytotoxicity of busulfan and4-hydroperoxycyclophosphamide in AML cells. Exp Hematol 2018; 67:49-59.e1. [PMID: 30102945 DOI: 10.1016/j.exphem.2018.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/23/2018] [Accepted: 08/05/2018] [Indexed: 01/09/2023]
Abstract
The combination of the DNA-alkylating agents busulfan (Bu) and cyclophosphamide is the most commonly used myeloablative pretransplantation conditioning therapy for myeloid leukemias. However, it is associated with significant nonrelapse mortality, which prohibits dose escalation to control relapse. We hypothesized that combining these two drugs with an epigenetic modifier would increase antileukemic efficacy without jeopardizing patient safety. A preclinical study was performed to determine the synergistic cytotoxicity of Bu, 4-hydroperoxycyclophosphamide (4HC), and the hypomethylating agent decitabine (DAC) in human acute myeloid leukemia (AML) cell lines. Exposure of KBM3/Bu2506 (P53-null) and OCI-AML3 (P53-wild-type) cells to Bu+4HC inhibited cell proliferation by ∼35-39%; addition of DAC increased the inhibition to ∼60-62%. The observed synergistic interactions correlated with DNA damage response activation, increased the production of reactive oxygen species, and decreased mitochondrial membrane potential, release of mitochondrial proapoptotic proteins into the cytoplasm, and induction of caspase-dependent programmed cell death. The Bu+4HC+DAC combination further caused chromatin trapping of DNMT1 with a concomitant increase in DNA damage. In contrast, FMS-like tyrosine kinase 3 internal tandem duplications (FLT3-ITD)-positive AML cell lines were not sensitized to Bu+4HC by inclusion of DAC; addition of the FLT3 kinase inhibitor sorafenib sensitized the FLT3-ITD-positive MV4-11 and MOLM13 cell lines to the triple drug combination by inhibiting the FLT3 signal transduction pathway. Our results therefore provide a rationale for the development of personalized conditioning therapy for patients with P53-mutated and FLT3-ITD-positive AML.
Collapse
Affiliation(s)
- Benigno C Valdez
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| | - Xiaowen Tang
- Department of Hematology, The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, Suzhou, China
| | - Yang Li
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - David Murray
- Department of Experimental Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada T6G 1Z2
| | - Yan Liu
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Uday Popat
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Borje S Andersson
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
19
|
Wong E, Davis JE, Grigg A, Szer J, Ritchie D. Strategies to enhance the graft versus tumour effect after allogeneic haematopoietic stem cell transplantation. Bone Marrow Transplant 2018; 54:175-189. [PMID: 29904127 DOI: 10.1038/s41409-018-0244-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 04/09/2018] [Accepted: 05/15/2018] [Indexed: 11/09/2022]
Abstract
Relapse of haematological malignancies after allogeneic haematopoietic stem cell transplant is a major cause of mortality. The immunological mechanisms that may lead to disease relapse may include immunological immaturity prior to reconstitution of the allogeneic immune system, tumour antigen downregulation or promotion of T-cell exhaustion by interactions with the tumour microenvironment. Current therapeutic strategies for post-transplant relapse are limited in their efficacy and alternative approaches are required. In this review, we discuss the mechanisms of T and NK-cell immune evasion that facilitate relapse of haematological malignancies after allogeneic stem cell transplantation, and explore emerging strategies to augment the allogeneic immune system in order to construct a more potent graft versus tumour response.
Collapse
Affiliation(s)
- Eric Wong
- Clinical Haematology and Bone Marrow Transplantation, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Victoria, Australia. .,Australian Cancer Research Foundation Translational Research Laboratory, Victoria, Australia. .,Department of Medicine, University of Melbourne, Victoria, Australia.
| | - Joanne E Davis
- Australian Cancer Research Foundation Translational Research Laboratory, Victoria, Australia.,Department of Medicine, University of Melbourne, Victoria, Australia
| | - Andrew Grigg
- Department of Medicine, University of Melbourne, Victoria, Australia.,Department of Clinical Haematology and Olivia Newton John Cancer Research Institute, Austin Hospital, Victoria, Australia
| | - Jeff Szer
- Clinical Haematology and Bone Marrow Transplantation, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Victoria, Australia.,Department of Medicine, University of Melbourne, Victoria, Australia
| | - David Ritchie
- Clinical Haematology and Bone Marrow Transplantation, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Victoria, Australia.,Australian Cancer Research Foundation Translational Research Laboratory, Victoria, Australia.,Department of Medicine, University of Melbourne, Victoria, Australia
| |
Collapse
|
20
|
Hochberg J, Zahler S, Geyer MB, Chen N, Krajewski J, Harrison L, Militano O, Ozkaynak MF, Cheerva AC, Talano J, Moore TB, Gillio AP, Walters MC, Baxter-Lowe LA, Hamby C, Cairo MS. The safety and efficacy of clofarabine in combination with high-dose cytarabine and total body irradiation myeloablative conditioning and allogeneic stem cell transplantation in children, adolescents, and young adults (CAYA) with poor-risk acute leukemia. Bone Marrow Transplant 2018; 54:226-235. [PMID: 29899571 DOI: 10.1038/s41409-018-0247-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 04/16/2018] [Accepted: 05/19/2018] [Indexed: 01/19/2023]
Abstract
Acute leukemias in children with CR3, refractory relapse, or induction failure (IF) have a poor prognosis. Clofarabine has single agent activity in relapsed leukemia and synergy with cytarabine. We sought to determine the safety and overall survival in a Phase I/II trial of conditioning with clofarabine (doses 40 - 52 mg/m2), cytarabine 1000 mg/m2, and 1200 cGy TBI followed by alloSCT in children, adolescents, and young adults with poor-risk leukemia. Thirty-seven patients; Age 12 years (1-22 years); ALL/AML: 34:3 (18 IF, 10 CR3, 13 refractory relapse); 15 related, 22 unrelated donors. Probabilities of neutrophil, platelet engraftment, acute GvHD, and chronic GvHD were 94%, 84%, 49%, and 30%, respectively. Probability of day 100 TRM was 8.1%. 2-year EFS (event free survival) and OS (overall survival) were 38.6% (CI95: 23-54%), and 41.3% (CI95: 25-57%). Multivariate analysis demonstrated overt disease at time of transplant (relative risk (RR) 3.65, CI95: 1.35-9.89, P = 0.011) and umbilical cord blood source (RR 2.17, CI95: 1.33-4.15, P = 0.019) to be predictors of worse EFS/OS. This novel myeloablative conditioning regimen followed by alloSCT is safe and well tolerated in CAYA with very poor-risk ALL or AML. Further investigation in CAYA with better risk ALL and AML undergoing alloSCT is warranted.
Collapse
Affiliation(s)
| | - Stacey Zahler
- Pediatric Institute, Cleveland Clinic Children's, Cleveland, Ohio, USA
| | - Mark B Geyer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nan Chen
- Departments of Pediatrics, Valhalla, NY, USA
| | - Jennifer Krajewski
- Department of Pediatrics, Hackensack University Medical Center, Hackensack, NJ, USA
| | | | | | | | | | - Julie Talano
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Theodore B Moore
- Department of Pediatrics, University of California at Los Angeles, Los Angeles, CA, USA
| | - Alfred P Gillio
- Department of Pediatrics, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Mark C Walters
- Department of Hematology/Oncology, Children's Hospital and Research Center of Oakland, Oakland, CA, USA
| | - Lee Ann Baxter-Lowe
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Carl Hamby
- Departments of Microbiology and Immunology, Valhalla, NY, USA
| | - Mitchell S Cairo
- Departments of Pediatrics, Valhalla, NY, USA. .,Departments of Microbiology and Immunology, Valhalla, NY, USA. .,Departments of Medicine, Valhalla, NY, USA. .,Departments of Pathology, Valhalla, NY, USA. .,Departments of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA.
| |
Collapse
|
21
|
Lowe KL, Mackall CL, Norry E, Amado R, Jakobsen BK, Binder G. Fludarabine and neurotoxicity in engineered T-cell therapy. Gene Ther 2018; 25:176-191. [DOI: 10.1038/s41434-018-0019-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/25/2018] [Accepted: 03/09/2018] [Indexed: 12/13/2022]
|
22
|
Haploidentical hematopoietic SCT using helical tomotherapy for total-body irradiation and targeted dose boost in patients with high-risk/refractory acute lymphoblastic leukemia. Bone Marrow Transplant 2018; 53:438-448. [PMID: 29330392 DOI: 10.1038/s41409-017-0049-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 09/12/2017] [Accepted: 11/17/2017] [Indexed: 11/08/2022]
Abstract
A novel conditioning regimen using helical tomotherapy (HT) was developed to deliver 10 Gy for total body irradiation (TBI) and simultaneously augment dose to 12 Gy for targeted dose boost to total marrow, central nervous system leukemia, and extramedullary disease sites in patients with high-risk or relapsed/refractory acute lymphoblastic leukemia (ALL) receiving haploidentical allogeneic hematopoietic stem cell transplantation (allo-HSCT). Fourteen patients were included, eight of these patients were in first complete remission (CR1), one was in CR2, one had a partial response and four patients had refractory disease at transplantation. The median delivered average dose was 11.395 Gy (range 10.06-12.17). The median planning target volume D95 was 8.2 Gy (range 7.52-9.01). The median delivered dose to skeleton bone with active bone marrow sites was 12.685 Gy (range 11.12-13.52). The results of this trial suggest that using HT TBI confers satisfactory immunosuppression and excellent eradication of malignant cells in patients with high-risk ALL undergoing allo-HSCT, especially in those with refractory ALL. After a median follow-up of 14.6 months (range 4-28), four patients experienced non-relapse mortality, ten patients are alive in durable CR including remission of extramedullary leukemic infiltration. One-year overall survival and disease-free survival rates post-transplantation were both 70.7%.
Collapse
|
23
|
Kebriaei P, Anasetti C, Zhang MJ, Wang HL, Aldoss I, de Lima M, Khoury HJ, Sandmaier BM, Horowitz MM, Artz A, Bejanyan N, Ciurea S, Lazarus HM, Gale RP, Litzow M, Bredeson C, Seftel MD, Pulsipher MA, Boelens JJ, Alvarnas J, Champlin R, Forman S, Pullarkat V, Weisdorf D, Marks DI. Intravenous Busulfan Compared with Total Body Irradiation Pretransplant Conditioning for Adults with Acute Lymphoblastic Leukemia. Biol Blood Marrow Transplant 2017; 24:726-733. [PMID: 29197676 DOI: 10.1016/j.bbmt.2017.11.025] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 11/22/2017] [Indexed: 01/22/2023]
Abstract
Total body irradiation (TBI) has been included in standard conditioning for acute lymphoblastic leukemia (ALL) before hematopoietic cell transplantation (HCT). Non-TBI regimens have incorporated busulfan (Bu) to decrease toxicity. This retrospective study analyzed TBI and Bu on outcomes of ALL patients 18-60 years old, in first or second complete remission (CR), undergoing HLA-compatible sibling, related, or unrelated donor HCT, who reported to the Center for International Blood and Marrow Transplant Research from 2005 to 2014. TBI plus etoposide (25%) or cyclophosphamide (75%) was used in 819 patients, and intravenous Bu plus fludarabine (41%), clofarabine (30%), cyclophosphamide (15%), or melphalan (13%) was used in 299 patients. Bu-containing regimens were analyzed together, since no significant differences for patient outcomes were noted between them. Bu patients were older, with better performance status; took longer to achieve first CR and receive HCT; were treated more recently; and were more likely to receive peripheral blood grafts, antithymocyte globulin, or tyrosine kinase inhibitors. With median follow-up of 3.6 years for Bu and 5.3 years for TBI, adjusted 3-year outcomes showed treatment-related mortality Bu 19% versus TBI 25% (P = .04); relapse Bu 37% versus TBI 28% (P = .007); disease-free survival (DFS) Bu 45% versus TBI 48% (P = .35); and overall survival (OS) Bu 57% versus TBI 53% (P = .35). In multivariate analysis, Bu patients had higher risk of relapse (relative risk, 1.46; 95% confidence interval, 1.15 to 1.85; P = .002) compared with TBI patients. Despite the higher relapse, Bu-containing conditioning led to similar OS and DFS following HCT for ALL.
Collapse
Affiliation(s)
- Partow Kebriaei
- Department of Stem Cell Transplantation, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Claudio Anasetti
- Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center Research Institute, Tampa, Florida
| | - Mei-Jie Zhang
- Division of Biostatistics, Institute for Health and Society, Medical College of Wisconsin, Milwaukee, Wisconsin; CIBMTR (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Hai-Lin Wang
- CIBMTR (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ibrahim Aldoss
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, California
| | - Marcos de Lima
- Department of Medicine, Seidman Cancer Center, University Hospitals Case Medical Center, Cleveland, Ohio
| | - H Jean Khoury
- Division of Hematology and Oncology, Emory University Hospital, Atlanta, Georgia
| | - Brenda M Sandmaier
- Division of Medical Oncology, University of Washington and Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Mary M Horowitz
- CIBMTR (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Andrew Artz
- Section of Hematology/Oncology, University of Chicago School of Medicine, Chicago, Illinois
| | - Nelli Bejanyan
- Division of Hematology, Oncology and Transplantation, University of Minnesota Medical Center, Minneapolis, Minnesota
| | - Stefan Ciurea
- Department of Stem Cell Transplantation, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hillard M Lazarus
- Department of Medicine, Seidman Cancer Center, University Hospitals Case Medical Center, Cleveland, Ohio
| | - Robert Peter Gale
- Hematology Research Centre, Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | - Mark Litzow
- Division of Hematology, Department of Internal Medicine, Mayo Clinic Rochester, Rochester, Minnesota
| | - Christopher Bredeson
- Ottawa Hospital Blood and Marrow Transplant Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Matthew D Seftel
- Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg, Manitoba, Canada
| | - Michael A Pulsipher
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Children's Hospital Los Angeles, USC Keck School of Medicine, Los Angeles, California
| | | | - Joseph Alvarnas
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, California
| | - Richard Champlin
- Department of Stem Cell Transplantation, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stephen Forman
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, California
| | - Vinod Pullarkat
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, California
| | - Daniel Weisdorf
- Division of Hematology, Oncology and Transplantation, University of Minnesota Medical Center, Minneapolis, Minnesota
| | - David I Marks
- Adult Bone Marrow Transplant, University Hospitals Bristol NHS Trust, Bristol, United Kingdom
| | | |
Collapse
|
24
|
Affiliation(s)
- Ibrahim Aldoss
- Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Anthony S. Stein
- Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| |
Collapse
|