1
|
Mohty R, Al Kadhimi Z, Kharfan-Dabaja M. Post-transplant cyclophosphamide or cell selection in haploidentical allogeneic hematopoietic cell transplantation? Hematology 2024; 29:2326384. [PMID: 38597828 DOI: 10.1080/16078454.2024.2326384] [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: 12/19/2023] [Accepted: 02/28/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND One major limitation for broader applicability of allogeneic hematopoietic cell transplantation (allo-HCT) in the past was the lack of HLA-matched histocompatible donors. Preclinical mouse studies using T-cell depleted haploidentical grafts led to an increased interest in the use of ex vivo T-cell depleted (TCD) haploidentical allo-HCT. TCD grafts through negative (T-cell depletion) or positive (CD34+ cell selection) techniques have been investigated to reduce the risk of graft-versus-host disease (GVHD) given the known implications of alloreactive T cells. A more practical approach to deplete alloreactive T cells in vivo using high doses of cyclophosphamide after allografting has proved to be feasible in overcoming the HLA barrier. Such approach has extended allo-HCT feasibility to patients for whom donors could not be found in the past. Nowadays, haploidentical donors represent a common donor source for patients in need of an allo-HCT. The broad application of haploidentical donors became possible by understanding the importance of depleting alloreactive donor T cells to facilitate engraftment and reduce incidence and severity of GVHD. These techniques involve ex vivo graft manipulation or in vivo utilization of pharmacologic agents, notably post-transplant cyclophosphamide (PTCy). DISCUSSION While acknowledging that no randomized controlled prospective studies have been yet conducted comparing TCD versus PTCy in haploidentical allo-HCT recipients, there are two advantages that would favor the PTCy, namely ease of application and lower cost. However, emerging data on adverse events associated with PTCy including, but not limited to cardiac associated toxicities or increased incidence of post-allograft infections, and others, are important to recognize.
Collapse
Affiliation(s)
- Razan Mohty
- Division of Hematology Oncology, Department of Medicine, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Zaid Al Kadhimi
- Division of Hematology Oncology, Department of Medicine, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Mohamed Kharfan-Dabaja
- Division of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
2
|
Möhn N, Grote-Levi L, Wattjes MP, Bonifacius A, Holzwart D, Hopfner F, Nay S, Tischer-Zimmermann S, Saßmann ML, Schwenkenbecher P, Sühs KW, Mahmoudi N, Warnke C, Zimmermann J, Hagin D, Goudeva L, Blasczyk R, Koch A, Maecker-Kolhoff B, Eiz-Vesper B, Höglinger G, Skripuletz T. Directly Isolated Allogeneic Virus-Specific T Cells in Progressive Multifocal Leukoencephalopathy. JAMA Neurol 2024:2824325. [PMID: 39374035 PMCID: PMC11459361 DOI: 10.1001/jamaneurol.2024.3324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 08/09/2024] [Indexed: 10/08/2024]
Abstract
Importance Progressive multifocal leukoencephalopathy (PML) is a life-threatening viral infection with no approved antiviral treatment. Objective To determine whether restoring the compromised immune system of patients with PML with directly isolated allogeneic virus-specific (DIAVIS) T cells is a promising therapeutic strategy, especially if other curative options are absent. Design, Setting, and Participants A retrospective case series of patients with PML who were treated with DIAVIS T cells was conducted between March 2020 and February 2022. T cells were isolated from healthy donors within 24 hours and targeted against the BK polyomavirus. Patients with PML were treated monocentrically. Eligibility for treatment with DIAVIS T cells was assessed for patients with confirmed PML, and exclusion criteria included stable PML disease and previous treatment with natalizumab. Exposure Fresh DIAVIS T cells were administered with a maximum dose of 2 × 104 CD3+ cells/kg body weight. Remaining T cells were cryopreserved in divided doses and administered in additional treatments approximately 2 and 6 weeks later. Main Outcomes and Measures Primary outcome measures were clinical response and survival of patients, compared with the outcomes of a historical reference group of PML cases receiving best supportive treatment (BST) and with recently published real-world data of patients with PML who were treated with immune checkpoint inhibition. Results The study cohort consisted of 28 patients (median [IQR] age, 60 [51-72] years; 20 male [71.4%]). Twenty-two patients (79%) treated with DIAVIS T cells showed response, resulting in significant clinical stabilization or improvement and a reduction in viral load. Six individuals (21%) were classified as nonresponders, deteriorated rapidly, and died, as did 2 other patients during a 12-month follow-up. Older age was the only predictor of a poor treatment response. Survival analysis revealed better 12-month survival rates (hazard ratio, 0.42; 95% CI, 0.24-0.73; P =.02) from diagnosis for patients treated with DIAVIS T cells (18 of 26 [69%]; 12-mo survival rate, 69%) compared with historical controls with BST (57 of 113 [50%]; 12-mo survival rate, including censored data, 45%). Conclusion and Relevance This case series of DIAVIS T-cell therapy in PML provides first class IV evidence suggesting efficacy to reduce mortality and improve functional outcome. Further prospective studies are required to confirm these results.
Collapse
Affiliation(s)
- Nora Möhn
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Lea Grote-Levi
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Mike P. Wattjes
- Department of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
- Department of Neuroradiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Agnes Bonifacius
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Dennis Holzwart
- Department of Biostatistics, Hannover Medical School, Hannover, Germany
| | - Franziska Hopfner
- Department of Neurology, LMU University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sandra Nay
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Sabine Tischer-Zimmermann
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | | | | | | | - Nima Mahmoudi
- Department of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
| | - Clemens Warnke
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | - David Hagin
- Allergy and Clinical Immunology Unit, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Lilia Goudeva
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Rainer Blasczyk
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Armin Koch
- Department of Biostatistics, Hannover Medical School, Hannover, Germany
| | - Britta Maecker-Kolhoff
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research, Hannover, Germany
| | - Britta Eiz-Vesper
- Department of Neuroradiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Infection Research, Hannover, Germany
| | - Günter Höglinger
- Department of Neurology, Hannover Medical School, Hannover, Germany
- Department of Neurology, LMU University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Thomas Skripuletz
- Department of Neurology, Hannover Medical School, Hannover, Germany
- Centre for Individualised Infection Medicine, Hannover, Germany
| |
Collapse
|
3
|
Chakraverty R. How Important Is Unrelated Donor Human Leukocyte Antigen Disparity in the Post-Transplant Cyclophosphamide Era? J Clin Oncol 2024; 42:3263-3265. [PMID: 39167734 DOI: 10.1200/jco-24-01303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 08/23/2024] Open
Affiliation(s)
- Ronjon Chakraverty
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
4
|
Pidala JA, Gooley TA, Luznik L, Blazar BR. Chronic graft-versus-host disease: unresolved complication or ancient history? Blood 2024; 144:1363-1373. [PMID: 39008818 PMCID: PMC11451335 DOI: 10.1182/blood.2023022735] [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: 04/25/2024] [Revised: 06/11/2024] [Accepted: 06/24/2024] [Indexed: 07/17/2024] Open
Abstract
ABSTRACT Chronic graft-versus-host disease (cGVHD) is associated with morbidity, mortality, impaired quality of life, prolonged immunosuppressive therapy, and infection risk after allogeneic hematopoietic cell transplantation (HCT). Major strides have occurred in the understanding of cGVHD biology; National Institutes of Health Consensus meetings have refined rigorous approaches to diagnosis, staging, and response criteria; major interventional trials have established standard benchmarks for treatment outcome; and 3 agents to date have been US Food and Drug Administration approved for treating corticosteroid-refractory cGVHD. Promising results from several recent trials have led some, but not others, to conclude that the risk of developing cGVHD is sufficiently low to be considered a major post-HCT complication of the past. We propose that it is time to critically examine the results of contemporary graft-versus-host disease (GVHD) prophylaxis regimens and discuss the state of the science and associated controversies in the spectrum of conclusions reached as to the risk of cGVHD. With these data, the current cGVHD incidence can be most precisely determined, and the present and future burden of cGVHD-affected patients can be accurately modeled. Through review of existing evidence, we highlight unresolved needs and opportunities to refine best GVHD prophylaxis or preemptive therapy approaches and optimize established cGVHD therapy, and make the argument that support of preclinical and clinical research is critical in improving patient outcomes.
Collapse
Affiliation(s)
- Joseph A. Pidala
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Ted A. Gooley
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Leo Luznik
- Section of Hematology and Oncology, Department of Internal Medicine, Baylor College of Medicine, Houston, TX
| | - Bruce R. Blazar
- Division of Blood and Marrow Transplant and Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| |
Collapse
|
5
|
Krakow EF, Brault M, Summers C, Cunningham TM, Biernacki MA, Black RG, Woodward KB, Vartanian N, Kanaan SB, Yeh AC, Dossa RG, Bar M, Cassaday RD, Dahlberg A, Till BG, Denker AE, Yeung CCS, Gooley TA, Maloney DG, Riddell SR, Greenberg PD, Chapuis AG, Newell EW, Furlan SN, Bleakley M. HA-1-targeted T-cell receptor T-cell therapy for recurrent leukemia after hematopoietic stem cell transplantation. Blood 2024; 144:1069-1082. [PMID: 38683966 PMCID: PMC11406181 DOI: 10.1182/blood.2024024105] [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: 01/29/2024] [Revised: 03/27/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
ABSTRACT Relapse is the leading cause of death after allogeneic hematopoietic stem cell transplantation (HCT) for leukemia. T cells engineered by gene transfer to express T cell receptors (TCR; TCR-T) specific for hematopoietic-restricted minor histocompatibility (H) antigens may provide a potent selective antileukemic effect post-HCT. We conducted a phase 1 clinical trial using a novel TCR-T product targeting the minor H antigen, HA-1, to treat or consolidate treatment of persistent or recurrent leukemia and myeloid neoplasms. The primary objective was to evaluate the feasibility and safety of administration of HA-1 TCR-T after HCT. CD8+ and CD4+ T cells expressing the HA-1 TCR and a CD8 coreceptor were successfully manufactured from HA-1-disparate HCT donors. One or more infusions of HA-1 TCR-T following lymphodepleting chemotherapy were administered to 9 HCT recipients who had developed disease recurrence after HCT. TCR-T cells expanded and persisted in vivo after adoptive transfer. No dose-limiting toxicities occurred. Although the study was not designed to assess efficacy, 4 patients achieved or maintained complete remissions following lymphodepletion and HA-1 TCR-T, with 1 patient still in remission at >2 years. Single-cell RNA sequencing of relapsing/progressive leukemia after TCR-T therapy identified upregulated molecules associated with T-cell dysfunction or cancer cell survival. HA-1 TCR-T therapy appears feasible and safe and shows preliminary signals of efficacy. This clinical trial was registered at ClinicalTrials.gov as #NCT03326921.
Collapse
Affiliation(s)
- Elizabeth F. Krakow
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Michelle Brault
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Corinne Summers
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Cancer and Blood Disorders Center, Seattle Children's Hospital, Seattle, WA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA
| | - Tanya M. Cunningham
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Melinda A. Biernacki
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - R. Graeme Black
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Kyle B. Woodward
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Nicole Vartanian
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Sami B. Kanaan
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Albert C. Yeh
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Robson G. Dossa
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Merav Bar
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Ryan D. Cassaday
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Ann Dahlberg
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Cancer and Blood Disorders Center, Seattle Children's Hospital, Seattle, WA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA
| | - Brian G. Till
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | | - Cecilia C. S. Yeung
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA
| | - Ted A. Gooley
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - David G. Maloney
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Stanley R. Riddell
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Philip D. Greenberg
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Immunology, University of Washington School of Medicine, Seattle, WA
| | - Aude G. Chapuis
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Evan W. Newell
- Vaccine and Infection Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Scott N. Furlan
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Cancer and Blood Disorders Center, Seattle Children's Hospital, Seattle, WA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA
| | - Marie Bleakley
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Cancer and Blood Disorders Center, Seattle Children's Hospital, Seattle, WA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
6
|
Holtzman NG, Curtis LM, Salit RB, Shaffer BC, Pirsl F, Ostojic A, Steinberg SM, Schulz E, Wilder JS, Hughes TE, Rose J, Memon S, Korngold R, Gea-Banacloche JC, Fowler DH, Hakim FT, Gress RE, Bishop MR, Pavletic SZ. High-dose alemtuzumab and cyclosporine vs tacrolimus, methotrexate, and sirolimus for chronic graft-versus-host disease prevention. Blood Adv 2024; 8:4294-4310. [PMID: 38669315 PMCID: PMC11372812 DOI: 10.1182/bloodadvances.2023010973] [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: 06/15/2023] [Revised: 02/05/2024] [Accepted: 02/26/2024] [Indexed: 04/28/2024] Open
Abstract
ABSTRACT Chronic graft-versus-host disease (cGVHD) remains a significant problem for patients after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Although in vivo lymphodepletion for cGVHD prophylaxis has been explored in the myeloablative setting, its effects after reduced-intensity conditioning (RIC) are not well described. Patients (N = 83) with hematologic malignancies underwent targeted lymphodepletion chemotherapy followed by a RIC allo-HSCT using peripheral blood stem cells from unrelated donors. Patients were randomized to 2 GVHD prophylaxis arms: alemtuzumab and cyclosporine (AC; n = 44) or tacrolimus, methotrexate, and sirolimus (TMS; n = 39), with the primary end point of cumulative incidence of severe cGVHD. The incidence of severe cGVHD was lower with AC vs TMS prophylaxis at 1- and 5-years (0% vs 10.3% and 4.5% vs 28.5%; overall, P = .0002), as well as any grade (P = .003) and moderate-severe (P < .0001) cGVHD. AC was associated with higher rates of grade 3 to 4 infections (P = .02) and relapse (52% vs 21%; P = .003) with no difference in 5-year GVHD-free-, relapse-free-, or overall survival. AC severely depleted naïve T-cell reconstitution, resulting in reduced T-cell receptor repertoire diversity, smaller populations of CD4Treg and CD8Tscm, but a higher ratio of Treg to naïve T-cells at 6 months. In summary, an alemtuzumab-based regimen successfully reduced the rate and severity of cGVHD after RIC allo-HSCT and resulted in a distinct immunomodulatory profile, which may have reduced cGVHD incidence and severity. However, increased infections and relapse resulted in a lack of survival benefit after long-term follow-up. This trial was registered at www.ClinicalTrials.gov as #NCT00520130.
Collapse
Affiliation(s)
- Noa G. Holtzman
- Immune Deficiency Cellular Therapy Program, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Lauren M. Curtis
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Ascension Maryland Saint Agnes Hospital, Baltimore, MD
| | - Rachel B. Salit
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Brian C. Shaffer
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Filip Pirsl
- Immune Deficiency Cellular Therapy Program, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Alen Ostojic
- Immune Deficiency Cellular Therapy Program, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Eduard Schulz
- Immune Deficiency Cellular Therapy Program, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jennifer S. Wilder
- Frederick National Laboratory for Cancer Research, Clinical Monitoring Research Program, Leidos Biomedical Research, Inc, Frederick, MD
| | - Thomas E. Hughes
- Department of Pharmacy, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Jeremy Rose
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Sarfraz Memon
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Robert Korngold
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ
| | - Juan C. Gea-Banacloche
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Daniel H. Fowler
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Rapa Therapeutics, Rockville, MD
| | - Frances T. Hakim
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ronald E. Gress
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Michael R. Bishop
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Department of Hematology/Oncology, David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL
| | - Steven Z. Pavletic
- Immune Deficiency Cellular Therapy Program, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
7
|
Appelbaum JS, Appelbaum FR, Percival ME. Second chances for secondary AML. Blood Adv 2024; 8:4221-4222. [PMID: 39136970 PMCID: PMC11372385 DOI: 10.1182/bloodadvances.2024013318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024] Open
Affiliation(s)
- Jacob S Appelbaum
- Division of Hematology/Oncology, Department of Medicine, University of Washington, Seattle, WA
- Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA
| | - Frederick R Appelbaum
- Division of Hematology/Oncology, Department of Medicine, University of Washington, Seattle, WA
- Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA
| | - Mary-Elizabeth Percival
- Division of Hematology/Oncology, Department of Medicine, University of Washington, Seattle, WA
- Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA
| |
Collapse
|
8
|
Gasior Kabat M, Li Y, Galán V, Mozo Y, Sisinni L, Bueno-Sánchez D, Corral D, Naik S, Echecopar C, Aguirre-Portolés C, Al-Akioui-Sanz K, De Paz R, Marcos A, Romero AB, Talleur A, Yuste VJ, Triplett B, Pérez-Martínez A. Safety and Efficacy of High-Dose Memory CD45RO + Donor Lymphocyte Infusion in Pediatric Recipients after Hematopoietic Stem Cell Transplantation. Cytotherapy 2024:S1465-3249(24)00799-0. [PMID: 39046390 DOI: 10.1016/j.jcyt.2024.07.007] [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: 04/16/2024] [Revised: 06/29/2024] [Accepted: 07/08/2024] [Indexed: 07/25/2024]
Abstract
Memory T selected cells (CD45RA-/RO+) as donor lymphocyte infusion are less capable of producing alloreactivity and graft versus host disease (GvHD) compared with naïve T cells. The objective of this study was to evaluate the safety and efficacy of high-dose memory (CD45RA-/RO+) donor lymphocyte infusion (mDLI) after allogeneic hematopoietic cell transplantation (HCT). Indications for mDLI were "as needed" and "as prophylactic regimen." Sixty-one children diagnosed with malignant (82%) and non-malignant diseases (18%) received 241 mDLIs. Patients received a median of three infusions (range 1‒13) of mDLI with a median infused dose of 1.35 × 107/kg CD45RO+ containing 8.96 × 106/kg CD3+CD45RO+ and 3.81 × 103/kg CD3+CD45RA+. De novo GvHD developed in 7 patients following 4% of the mDLI infusions. Among patients with GvHD before mDLI, this condition worsened following 6 infusions (11%) in the 3 patients with grade II-IV acute GvHD. A decrease in cytomegalovirus viral load followed 65% of mDLI infusions. Two-year overall survival (OS) for the total cohort was 64% (95% CI 57%‒72%). For patients receiving prophylactic mDLI, the two-year non-relapse mortality was 10% (95% CI 9%‒11%). In summary, high-dose mDLI is feasible and safe, with a relatively low risk of severe GvHD even in patients with active GvHD. Importantly, mDLI was associated with positive effects, including enhanced control of CMV viremia.
Collapse
Affiliation(s)
| | - Ying Li
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Victor Galán
- Pediatric Hematology-Oncology Department, La Paz University Hospital, Madrid, Spain
| | - Yasmina Mozo
- Pediatric Hematology-Oncology Department, La Paz University Hospital, Madrid, Spain
| | - Luisa Sisinni
- Pediatric Hematology-Oncology Department, La Paz University Hospital, Madrid, Spain
| | - David Bueno-Sánchez
- Pediatric Hematology-Oncology Department, La Paz University Hospital, Madrid, Spain
| | - Dolores Corral
- Pediatric Hematology-Oncology Department, La Paz University Hospital, Madrid, Spain
| | - Swati Naik
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Carlos Echecopar
- Pediatric Hematology-Oncology Department, La Paz University Hospital, Madrid, Spain
| | | | - Karima Al-Akioui-Sanz
- CIBERER-ISCIII, IdiPAZ-CNIO Pediatric OncoHematology Clinical Research Unit, Madrid, Spain
| | - Raquel De Paz
- Hematology Department, La Paz University Hospital, Madrid, Spain
| | - Antonio Marcos
- Hematology Department, La Paz University Hospital, Madrid, Spain
| | - Ana Belén Romero
- Hematology Department, La Paz University Hospital, Madrid, Spain
| | - Aimee Talleur
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Brandon Triplett
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Antonio Pérez-Martínez
- Pediatric Hematology-Oncology Department, La Paz University Hospital, Madrid, Spain; Pediatric Department. Autonomous University of Madrid College of Medicine, Madrid, Spain; CIBERER-ISCIII, IdiPAZ-CNIO Pediatric OncoHematology Clinical Research Unit, Madrid, Spain.
| |
Collapse
|
9
|
Guan L, Sun Y, Si Y, Yan Q, Han Z, Liu Y, Han T. A strategy to reconstitute immunity without GVHD via adoptive allogeneic Tscm therapy. Front Immunol 2024; 15:1367609. [PMID: 39035005 PMCID: PMC11259968 DOI: 10.3389/fimmu.2024.1367609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 06/19/2024] [Indexed: 07/23/2024] Open
Abstract
Introduction Adoption of allogeneic T cells directly supplements the number of T cells and rapidly induces T-cell immunity, which has good efficacy for treating some tumors and immunodeficiency diseases. However, poor adoptive T-cell engraftment and graft-versus-host disease (GVHD) limit the application of these methods. Alloreactive T-cell clones were eliminated from the donor T-cell repertoire, and the remaining T-cell clones were prepared as Tscm for T-cell adoptive treatment to reconstruct recipient T-cell immunity without GVHD. Methods The subjects in this study included three different strains of mice. Lymphocytes from mice (C57BL/6) were used as the donor T-cell repertoire, from which the Tscm allo-reactive T cell clone was depleted (ATD-Tscm). This was confirmed by showing that the Tscm was not responsive to the alloantigen of the recipient (BALB/c). To prepare ATD-Tscm cells, we used recipient lymphocytes as a simulator, and coculture of mouse and recipient lymphocytes was carried out for 7 days. Sorting of non-proliferative cells ensured that the prepared Tscm cells were nonresponsive. The sorted lymphocytes underwent further expansion by treatment with TWS119 and cytokines for an additional 10 days, after which the number of ATD-Tscm cells increased. The prepared Tscm cells were transferred into recipient mice to observe immune reconstitution and GVHD incidence. Results Our protocol began with the use of 1×107 donor lymphocytes and resulted in 1 ×107 ATD-Tscm cells after 17 days of preparation. The prepared ATD-Tscm cells exhibited a nonresponse upon restimulation of the recipient lymphocytes. Importantly, the prepared ATD-Tscm cells were able to bind long and reconstitute other T-cell subsets in vivo, effectively recognizing and answering the "foreign" antigen without causing GVHD after they were transferred into the recipients. Discussion Our strategy was succeeded to prepare ATD-Tscm cells from the donor T-cell repertoire. The prepared ATD-Tscm cells were able to reconstitute the immune system and prevent GVHD after transferred to the recipients. This study provides a good reference for generating ATD-Tscm for T-cell adoptive immunotherapy.
Collapse
Affiliation(s)
- Liping Guan
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yunqin Sun
- Clinical Department, Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Yanli Si
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Qingya Yan
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key laboratory for Molecular Oncology, Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, China
| | - Ziyu Han
- Xinxiang Key laboratory for Molecular Oncology, Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, China
| | - Youxun Liu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Tao Han
- Xinxiang Key laboratory for Molecular Oncology, Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
10
|
Naik S, Li Y, Talleur AC, Selukar S, Ashcraft E, Cheng C, Madden RM, Mamcarz E, Qudeimat A, Sharma A, Srinivasan A, Suliman AY, Epperly R, Obeng EA, Velasquez MP, Langfitt D, Schell S, Métais JY, Arnold PY, Hijano DR, Maron G, Merchant TE, Akel S, Leung W, Gottschalk S, Triplett BM. Memory T-cell enriched haploidentical transplantation with NK cell addback results in promising long-term outcomes: a phase II trial. J Hematol Oncol 2024; 17:50. [PMID: 38937803 PMCID: PMC11212178 DOI: 10.1186/s13045-024-01567-0] [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: 05/06/2024] [Accepted: 06/13/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Relapse remains a challenge after transplantation in pediatric patients with hematological malignancies. Myeloablative regimens used for disease control are associated with acute and long-term adverse effects. We used a CD45RA-depleted haploidentical graft for adoptive transfer of memory T cells combined with NK-cell addback and hypothesized that maximizing the graft-versus-leukemia (GVL) effect might allow for reduction in intensity of conditioning regimen. METHODS In this phase II clinical trial (NCT01807611), 72 patients with hematological malignancies (complete remission (CR)1: 25, ≥ CR2: 28, refractory disease: 19) received haploidentical CD34 + enriched and CD45RA-depleted hematopoietic progenitor cell grafts followed by NK-cell infusion. Conditioning included fludarabine, thiotepa, melphalan, cyclophosphamide, total lymphoid irradiation, and graft-versus-host disease (GVHD) prophylaxis consisted of a short-course sirolimus or mycophenolate mofetil without serotherapy. RESULTS The 3-year overall survival (OS) and event-free-survival (EFS) for patients in CR1 were 92% (95% CI:72-98) and 88% (95% CI: 67-96); ≥ CR2 were 81% (95% CI: 61-92) and 68% (95% CI: 47-82) and refractory disease were 32% (95% CI: 11-54) and 20% (95% CI: 6-40). The 3-year EFS for all patients in morphological CR was 77% (95% CI: 64-87) with no difference amongst recipients with or without minimal residual disease (P = 0.2992). Immune reconstitution was rapid, with mean CD3 and CD4 T-cell counts of 410/μL and 140/μL at day + 30. Cumulative incidence of acute GVHD and chronic GVHD was 36% and 26% but most patients with acute GVHD recovered rapidly with therapy. Lower rates of grade III-IV acute GVHD were observed with NK-cell alloreactive donors (P = 0.004), and higher rates of moderate/severe chronic GVHD occurred with maternal donors (P = 0.035). CONCLUSION The combination of a CD45RA-depleted graft and NK-cell addback led to robust immune reconstitution maximizing the GVL effect and allowed for use of a submyeloablative, TBI-free conditioning regimen that was associated with excellent EFS resulting in promising long-term outcomes in this high-risk population. The trial is registered at ClinicalTrials.gov (NCT01807611).
Collapse
Affiliation(s)
- Swati Naik
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA.
| | - Ying Li
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Aimee C Talleur
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Subodh Selukar
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Emily Ashcraft
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Cheng Cheng
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Renee M Madden
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ewelina Mamcarz
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Amr Qudeimat
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Akshay Sharma
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ashok Srinivasan
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ali Y Suliman
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Rebecca Epperly
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Esther A Obeng
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - M Paulina Velasquez
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Deanna Langfitt
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Sarah Schell
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Jean-Yves Métais
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Paula Y Arnold
- Department of Pathology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Diego R Hijano
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Gabriela Maron
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Thomas E Merchant
- Department of Radiation Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Salem Akel
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Wing Leung
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Brandon M Triplett
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
11
|
Wang Y, Ullah MA, Waltner OG, Bhise SS, Ensbey KS, Schmidt CR, Legg SR, Sekiguchi T, Nelson EL, Kuns RD, Nemychenkov NS, Atilla E, Yeh AC, Takahashi S, Boiko JR, Varelias A, Blazar BR, Koyama M, Minnie SA, Clouston AD, Furlan SN, Zhang P, Hill GR. Calcineurin inhibition rescues alloantigen-specific central memory T cell subsets that promote chronic GVHD. J Clin Invest 2024; 134:e170125. [PMID: 38828727 PMCID: PMC11142741 DOI: 10.1172/jci170125] [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: 03/02/2023] [Accepted: 04/09/2024] [Indexed: 06/05/2024] Open
Abstract
Calcineurin inhibitors (CNIs) constitute the backbone of modern acute graft-versus-host disease (aGVHD) prophylaxis regimens but have limited efficacy in the prevention and treatment of chronic GVHD (cGVHD). We investigated the effect of CNIs on immune tolerance after stem cell transplantation with discovery-based single-cell gene expression and T cell receptor (TCR) assays of clonal immunity in tandem with traditional protein-based approaches and preclinical modeling. While cyclosporin and tacrolimus suppressed the clonal expansion of CD8+ T cells during GVHD, alloreactive CD4+ T cell clusters were preferentially expanded. Moreover, CNIs mediated reversible dose-dependent suppression of T cell activation and all stages of donor T cell exhaustion. Critically, CNIs promoted the expansion of both polyclonal and TCR-specific alloreactive central memory CD4+ T cells (TCM) with high self-renewal capacity that mediated cGVHD following drug withdrawal. In contrast to posttransplant cyclophosphamide (PT-Cy), CSA was ineffective in eliminating IL-17A-secreting alloreactive T cell clones that play an important role in the pathogenesis of cGVHD. Collectively, we have shown that, although CNIs attenuate aGVHD, they paradoxically rescue alloantigen-specific TCM, especially within the CD4+ compartment in lymphoid and GVHD target tissues, thus predisposing patients to cGVHD. These data provide further evidence to caution against CNI-based immune suppression without concurrent approaches that eliminate alloreactive T cell clones.
Collapse
Affiliation(s)
- Yewei Wang
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Md Ashik Ullah
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Olivia G. Waltner
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Shruti S. Bhise
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Kathleen S. Ensbey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Christine R. Schmidt
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Samuel R.W. Legg
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Tomoko Sekiguchi
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Ethan L. Nelson
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Rachel D. Kuns
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Nicole S. Nemychenkov
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Erden Atilla
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Albert C. Yeh
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Shuichiro Takahashi
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Julie R. Boiko
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Antiopi Varelias
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Bruce R. Blazar
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Motoko Koyama
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Simone A. Minnie
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | | | - Scott N. Furlan
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Pediatrics and
| | - Ping Zhang
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Geoffrey R. Hill
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Division of Medical Oncology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
12
|
Belbachir S, Abraham A, Sharma A, Prockop S, DeZern AE, Bonfim C, Bidgoli A, Li J, Ruggeri A, Bertaina A, Boelens JJ, Purtill D. Engineering the best transplant outcome for high-risk acute myeloid leukemia: the donor, the graft and beyond. Cytotherapy 2024; 26:546-555. [PMID: 38054912 DOI: 10.1016/j.jcyt.2023.11.004] [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: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 12/07/2023]
Abstract
Allogeneic hemopoietic cell transplantation remains the goal of therapy for high-risk acute myeloid leukemia (AML). However, treatment failure in the form of leukemia relapse or severe graft-versus-host disease remains a critical area of unmet need. Recently, significant progress has been made in the cell therapy-based interventions both before and after transplant. In this review, the Stem Cell Engineering Committee of the International Society for Cell and Gene Therapy summarizes the literature regarding the identification of high risk in AML, treatment approaches before transplant, optimal transplant platforms and measures that may be taken after transplant to ideally prevent, or, if need be, treat AML relapse. Although some strategies remain in the early phases of clinical investigation, they are built on progress in pre-clinical research and cellular engineering techniques that are already improving outcomes for children and adults with high-risk malignancies.
Collapse
Affiliation(s)
- Safia Belbachir
- Haematology Department, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - Allistair Abraham
- Center for Cancer and Immunology Research, CETI, Children's National Hospital, Washington, District of Columbia, USA
| | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Susan Prockop
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts USA
| | - Amy E DeZern
- Bone Marrow Failure and MDS Program, John Hopkins Medicine, Baltimore, Maryland, USA
| | - Carmem Bonfim
- Pediatric Blood and Marrow Transplantation Division/Instituto de Pesquisa Pele Pequeno Principe Research/Faculdades Pequeno Príncipe, Curitiba, Brazil
| | - Alan Bidgoli
- Division of Blood and Marrow Transplantation, Children's Healthcare of Atlanta, Aflac Blood and Cancer Disorders Center, Emory University, Atlanta, Georgia, USA
| | - Jinjing Li
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
| | | | - Alice Bertaina
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Jaap Jan Boelens
- Stem Cell Transplantation and Cellular Therapies, Memorial Sloan Kettering Cancer Center, and Department of Pediatrics, Weill Cornell Medical College of Cornell University, New York, New York, USA
| | - Duncan Purtill
- Haematology Department, Fiona Stanley Hospital, Perth, Western Australia, Australia; PathWest Laboratory Medicine, Perth, Western Australia, Australia.
| |
Collapse
|
13
|
Maurer K, Antin JH. The graft versus leukemia effect: donor lymphocyte infusions and cellular therapy. Front Immunol 2024; 15:1328858. [PMID: 38558819 PMCID: PMC10978651 DOI: 10.3389/fimmu.2024.1328858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is a potentially curative therapy for many hematologic malignancies as well as non-malignant conditions. Part of the curative basis underlying HSCT for hematologic malignancies relies upon induction of the graft versus leukemia (GVL) effect in which donor immune cells recognize and eliminate residual malignant cells within the recipient, thereby maintaining remission. GVL is a clinically evident phenomenon; however, specific cell types responsible for inducing this effect and molecular mechanisms involved remain largely undefined. One of the best examples of GVL is observed after donor lymphocyte infusions (DLI), an established therapy for relapsed disease or incipient/anticipated relapse. DLI involves infusion of peripheral blood lymphocytes from the original HSCT donor into the recipient. Sustained remission can be observed in 20-80% of patients treated with DLI depending upon the underlying disease and the intrinsic burden of targeted cells. In this review, we will discuss current knowledge about mechanisms of GVL after DLI, experimental strategies for augmenting GVL by manipulation of DLI (e.g. neoantigen vaccination, specific cell type selection/depletion) and research outlook for improving DLI and cellular immunotherapies for hematologic malignancies through better molecular definition of the GVL effect.
Collapse
Affiliation(s)
| | - Joseph H. Antin
- Division of Hematologic Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
14
|
Di Ianni M, Liberatore C, Santoro N, Ranalli P, Guardalupi F, Corradi G, Villanova I, Di Francesco B, Lattanzio S, Passeri C, Lanuti P, Accorsi P. Cellular Strategies for Separating GvHD from GvL in Haploidentical Transplantation. Cells 2024; 13:134. [PMID: 38247827 PMCID: PMC10814899 DOI: 10.3390/cells13020134] [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: 12/02/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
GvHD still remains, despite the continuous improvement of transplantation platforms, a fearful complication of transplantation from allogeneic donors. Being able to separate GvHD from GvL represents the greatest challenge in the allogeneic transplant setting. This may be possible through continuous improvement of cell therapy techniques. In this review, current cell therapies are taken into consideration, which are based on the use of TCR alpha/beta depletion, CD45RA depletion, T regulatory cell enrichment, NK-cell-based immunotherapies, and suicide gene therapies in order to prevent GvHD and maximally amplify the GvL effect in the setting of haploidentical transplantation.
Collapse
Affiliation(s)
- Mauro Di Ianni
- Hematology Unit, Pescara Hospital, 65124 Pescara, Italy; (C.L.); (N.S.); (P.R.)
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, 66100 Chieti, Italy; (F.G.); (G.C.); (S.L.); (P.L.)
- Center for Advanced Studies and Technology (CAST), University of Chieti-Pescara, 66100 Chieti, Italy
| | - Carmine Liberatore
- Hematology Unit, Pescara Hospital, 65124 Pescara, Italy; (C.L.); (N.S.); (P.R.)
| | - Nicole Santoro
- Hematology Unit, Pescara Hospital, 65124 Pescara, Italy; (C.L.); (N.S.); (P.R.)
| | - Paola Ranalli
- Hematology Unit, Pescara Hospital, 65124 Pescara, Italy; (C.L.); (N.S.); (P.R.)
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, 66100 Chieti, Italy; (F.G.); (G.C.); (S.L.); (P.L.)
- Center for Advanced Studies and Technology (CAST), University of Chieti-Pescara, 66100 Chieti, Italy
| | - Francesco Guardalupi
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, 66100 Chieti, Italy; (F.G.); (G.C.); (S.L.); (P.L.)
- Center for Advanced Studies and Technology (CAST), University of Chieti-Pescara, 66100 Chieti, Italy
| | - Giulia Corradi
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, 66100 Chieti, Italy; (F.G.); (G.C.); (S.L.); (P.L.)
- Center for Advanced Studies and Technology (CAST), University of Chieti-Pescara, 66100 Chieti, Italy
| | - Ida Villanova
- Blood Bank Unit, Pescara Hospital, 65124 Pescara, Italy; (I.V.); (B.D.F.); (C.P.); (P.A.)
| | - Barbara Di Francesco
- Blood Bank Unit, Pescara Hospital, 65124 Pescara, Italy; (I.V.); (B.D.F.); (C.P.); (P.A.)
| | - Stefano Lattanzio
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, 66100 Chieti, Italy; (F.G.); (G.C.); (S.L.); (P.L.)
- Center for Advanced Studies and Technology (CAST), University of Chieti-Pescara, 66100 Chieti, Italy
| | - Cecilia Passeri
- Blood Bank Unit, Pescara Hospital, 65124 Pescara, Italy; (I.V.); (B.D.F.); (C.P.); (P.A.)
| | - Paola Lanuti
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, 66100 Chieti, Italy; (F.G.); (G.C.); (S.L.); (P.L.)
- Center for Advanced Studies and Technology (CAST), University of Chieti-Pescara, 66100 Chieti, Italy
| | - Patrizia Accorsi
- Blood Bank Unit, Pescara Hospital, 65124 Pescara, Italy; (I.V.); (B.D.F.); (C.P.); (P.A.)
| |
Collapse
|
15
|
Ferreras C, Hernández-Blanco C, Martín-Quirós A, Al-Akioui-Sanz K, Mora-Rillo M, Ibáñez F, Díaz-Almirón M, Cano-Ochando J, Lozano-Ojalvo D, Jiménez-González M, Goterris R, Sánchez-Zapardiel E, de Paz R, Guerra-García P, Queiruga-Parada J, Molina P, Briones ML, Ruz-Caracuel B, Borobia AM, Carcas AJ, Planelles D, Vicario JL, Moreno MÁ, Balas A, Llano M, Llorente A, Del Balzo Á, Cañada C, García MÁ, Calvin ME, Arenas I, Pérez de Diego R, Eguizábal C, Soria B, Solano C, Pérez-Martínez A. Results of phase 2 randomized multi-center study to evaluate the safety and efficacy of infusion of memory T cells as adoptive therapy in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pneumonia and/or lymphopenia (RELEASE NCT04578210). Cytotherapy 2024; 26:25-35. [PMID: 37897472 DOI: 10.1016/j.jcyt.2023.10.002] [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: 06/11/2023] [Revised: 09/05/2023] [Accepted: 10/05/2023] [Indexed: 10/30/2023]
Abstract
BACKGROUND AIMS There are currently no effective anti-viral treatments for coronavirus disease 2019 (COVID-19)-hospitalized patients with hypoxemia. Lymphopenia is a biomarker of disease severity usually present in patients who are hospitalized. Approaches to increasing lymphocytes exerting an anti-viral effect must be considered to treat these patients. Following our phase 1 study, we performed a phase 2 randomized multicenter clinical trial in which we evaluated the efficacy of the infusion of allogeneic off-the-shelf CD45RA- memory T cells containing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific T cells from convalescent donors plus the standard of care (SoC) versus just the SoC treatment. METHODS Eighty-four patients were enrolled in three Spanish centers. The patients were randomized into the infusion of 1 × 106/kg CD45RA- memory T cells or the SoC. We selected four unvaccinated donors based on the expression of interferon gamma SARS-CoV-2-specific response within the CD45RA- memory T cells and the most frequent human leukocyte antigen typing in the Spanish population. RESULTS We analyzed data from 81 patients. The primary outcome for recovery, defined as the proportion of participants in each group with normalization of fever, oxygen saturation sustained for at least 24 hours and lymphopenia recovery through day 14 or at discharge, was met for the experimental arm. We also observed faster lymphocyte recovery in the experimental group. We did not observe any treatment-related adverse events. CONCLUSIONS Adoptive cell therapy with off-the-shelf CD45RA- memory T cells containing SAR-CoV-2-specific T cells is safe, effective and accelerates lymphocyte recovery of patients with COVID-19 pneumonia and/or lymphopenia. TRIAL REGISTRATION NCT04578210.
Collapse
Affiliation(s)
- Cristina Ferreras
- Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain
| | - Clara Hernández-Blanco
- Internal Medicine Department, Hospital de Emergencias Enfermera Isabel Zendal, Madrid, Spain
| | | | - Karima Al-Akioui-Sanz
- Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain
| | - Marta Mora-Rillo
- Infectious Diseases Unit, Internal Medicine Department, University Hospital La Paz, Hospital La Paz Institute for Health Research, IdiPAZ, Consorcio Centro de Investigación Biomédica en Red CIBER-Infec, Madrid, Spain
| | - Fátima Ibáñez
- Internal Medicine Department, Hospital Puerta de Hierro, Madrid, Spain
| | | | - Jordi Cano-Ochando
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Centro Nacional de Microbiologia, Instituto de Salud Carlos III, 28220 Madrid, Spain; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Daniel Lozano-Ojalvo
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - María Jiménez-González
- Infectious Diseases Unit, Internal Medicine Department, University Hospital La Paz, Hospital La Paz Institute for Health Research, IdiPAZ, Consorcio Centro de Investigación Biomédica en Red CIBER-Infec, Madrid, Spain; Clinical Trials Unit (UCICEC) at Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain
| | - Rosa Goterris
- Hematology Department, Hospital Clinico Universitario, Valencia, Spain
| | | | - Raquel de Paz
- Hematology Department, University Hospital La Paz, Madrid, Spain
| | - Pilar Guerra-García
- Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain; Pediatric Hemato-Oncology Department, University Hospital La Paz, Madrid, Spain
| | | | - Pablo Molina
- Clinical Pharmacology Department, University Hospital La Paz, Madrid, Spain
| | | | - Beatriz Ruz-Caracuel
- Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain
| | - Alberto M Borobia
- Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain; Clinical Trials Unit (UCICEC) at Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain; Clinical Pharmacology Department, University Hospital La Paz, Madrid, Spain
| | - Antonio J Carcas
- Clinical Trials Unit (UCICEC) at Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain; Clinical Pharmacology Department, University Hospital La Paz, Madrid, Spain; Faculty of Medicine Universidad Autónoma de Madrid, Madrid, Spain
| | - Dolores Planelles
- Department of Histocompatibility, Centro de Transfusión de la Comunidad Valenciana, Valencia, Spain
| | - José Luis Vicario
- Histocompatibility, Centro de Transfusión de la Comunidad de Madrid, Madrid, Spain
| | - Miguel Ángel Moreno
- Histocompatibility, Centro de Transfusión de la Comunidad de Madrid, Madrid, Spain
| | - Antonio Balas
- Histocompatibility, Centro de Transfusión de la Comunidad de Madrid, Madrid, Spain
| | - Marta Llano
- Infectious Diseases Department, Hospital de Emergencias Enfermera Isabel Zendal, Madrid, Spain
| | - Andrea Llorente
- Infectious Diseases Department, Hospital de Emergencias Enfermera Isabel Zendal, Madrid, Spain
| | - Álvaro Del Balzo
- Emergency Unit, Internal Medicine Department, University Hospital La Paz, Madrid, Spain
| | - Carlos Cañada
- Emergency Unit, Internal Medicine Department, University Hospital La Paz, Madrid, Spain
| | - Miguel Ángel García
- Emergency Unit, Internal Medicine Department, University Hospital La Paz, Madrid, Spain
| | - María Elena Calvin
- Emergency Unit, Internal Medicine Department, University Hospital La Paz, Madrid, Spain
| | - Isabel Arenas
- Emergency Unit, Internal Medicine Department, University Hospital La Paz, Madrid, Spain
| | - Rebeca Pérez de Diego
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid, Spain; Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid, Spain; Interdepartmental Group of Immunodeficiencies, Madrid, Spain
| | - Cristina Eguizábal
- Research Unit, Basque Centre for Blood Transfusion and Human Tissues, Osakidetza, Bizkaia, Spain; Biocruces Bizkaia Health Research Institute, Bizkaia, Spain
| | - Bernat Soria
- Health Research Institute-ISABIAL, Alicante University Hospital and Institute of Bioengineering, Miguel Hernández University, Alicante, Spain; University Pablo de Olavide, Sevilla, Spain
| | - Carlos Solano
- Hematology Department, Hospital Clinico Universitario, Valencia, Spain; Department of Medicine, University of Valencia, Valencia, Spain
| | - Antonio Pérez-Martínez
- Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain; Pediatric Hemato-Oncology Department, University Hospital La Paz, Madrid, Spain; Faculty of Medicine Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
16
|
Watkins B, Qayed M. Novel approaches to acute graft-versus-host disease prevention. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:155-163. [PMID: 38066861 PMCID: PMC10727007 DOI: 10.1182/hematology.2023000426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
The field of graft-versus-host disease (GvHD) has experienced significant growth, with increased number of clinical trials and the approval of several agents by the US Food and Drug Administration for both acute and chronic GvHD treatment. In addition, the development of prognostic biomarker algorithms has enabled risk stratification in acute GvHD. However, prevention remains the cornerstone of GvHD management. Notable recent changes include the expansion of donor options with the increased use of haploidentical donor and unrelated donor transplantation, the development of ex vivo selective T-cell depletion strategies, recent approval by the Food and Drug Administration of abatacept for GvHD prevention, and the application of posttransplant cyclophosphamide in matched and mismatched donor settings. In this article, we review the results of recent clinical trials in GvHD prophylaxis and discuss the changes in clinical practice and promising emerging strategies driving the field forward.
Collapse
Affiliation(s)
- Benjamin Watkins
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, and Emory University, Atlanta, GA
| | - Muna Qayed
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, and Emory University, Atlanta, GA
| |
Collapse
|
17
|
Amanam I, Otoukesh S, Al Malki MM, Salhotra A. Chronic GVHD: review advances in prevention, novel endpoints, and targeted strategies. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:164-170. [PMID: 38066845 PMCID: PMC10727045 DOI: 10.1182/hematology.2023000427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is a curative therapy for many malignant and non-malignant hematologic disorders. Chronic graft-versus-host (cGVHD) disease remains a significant hurdle for long-term survival in patients post allo-HCT, and it remains the leading cause of late non-relapse mortality. The risk factors for development of cGVHD include degree of human leukocyte antigen (HLA) disparity, increasing recipient age, use of peripheral blood stem cells as a source, myeloablative conditioning regimens, prior acute GVHD (aGVHD), and female donor to male recipient. Our biological understanding of cGVHD is mostly derived from transplantation mouse models and patient data. There are three distinct phases in the development of cGVHD. Approaches to prevent GVHD include pharmacologic strategies such as calcineurin inhibitors (cyclosporine, tacrolimus) combined with methotrexate or mTOR inhibitors (sirolimus), and IMP dehydrogenase inhibitors (mycophenolate mofetil). Increasingly, posttransplant cyclophosphamide is emerging as a promising strategy for GVCHD prevention especially in a setting of reduced intensity conditioning. Other approaches include serotherapy (ATG, Campath) and graft manipulation strategies. A significant obstacle to evaluating the response of novel GVHD-directed therapies has been standardized response assessments. This has functioned as a barrier to designing and interpreting clinical trials that are structured around the treatment of cGVHD. Novel endpoints including failure-free survival, Graft-versus-host disease-free, relapse-free survival (GRFS), and current GVHD-free, relapse-free survival (CGRFS) may create a clearer picture for post-HCT outcomes. Targeted therapies including Bruton's tyrosine kinase inhibition, JAK1/2 inhibition, and ROCK2 inhibitors have improved cGVHD therapy, especially in the steroid refractory setting. Continued improvement in prophylactic strategies for cGVHD, identification of accurate cGVHD treatment endpoints, and access to novel therapeutic agents are expected to improve cGVHD outcomes.
Collapse
|
18
|
Teshima T, Hashimoto D. Separation of GVL from GVHD -location, location, location. Front Immunol 2023; 14:1296663. [PMID: 38116007 PMCID: PMC10728488 DOI: 10.3389/fimmu.2023.1296663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/21/2023] [Indexed: 12/21/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation (HCT) is a curative therapy for various hematologic malignancies. However, alloimmune response is a double-edged sword that mediates both beneficial graft-versus-leukemia (GVL) effects and harmful graft-versus-host disease (GVHD). Separation of GVL effects from GVHD has been a topic of intense research to improve transplant outcomes, but reliable clinical strategies have not yet been established. Target tissues of acute GVHD are the skin, liver, and intestine, while leukemic stem cells reside in the bone marrow. Tissue specific effector T-cell migration is determined by a combination of inflammatory and chemotactic signals that interact with specific receptors on T cells. Specific inhibition of donor T cell migration to GVHD target tissues while preserving migration to the bone marrow may represent a novel strategy to separate GVL from GVHD. Furthermore, tissue specific GVHD therapy, promoting tissue tolerance, and targeting of the tumor immune microenvironment may also help to separate GVHD and GVL.
Collapse
Affiliation(s)
- Takanori Teshima
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | | |
Collapse
|
19
|
Miranda M, Gianfreda F, Carlotta D, Armati S, Barlattani A, Bollero P. Oral Manifestations of Graft vs. Host Disease: A Comprehensive Review for Best Practice in Dentistry. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1937. [PMID: 38003987 PMCID: PMC10673022 DOI: 10.3390/medicina59111937] [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: 09/28/2023] [Revised: 10/15/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023]
Abstract
Graft-versus-host disease (GVHD) is a complication of hematopoietic stem cell transplantation (HSCT). GVHD may also develop following solid transplants or blood transfusions if white blood cells are transferred. GVHD affects multiple organs, including the oral tissues. This pictorial review provides a background of GVHD to dental practitioners, describes the most common oral manifestations of GVHD, and highlights the main treatment modifications needed to deliver dental care to patients with GVHD. A narrative review enriched with clinical data was performed by searching the scientific literature for all articles regarding GVHD and oral manifestations/therapies. All articles without exclusion criteria, except animal tests, were included in the above review. Acute GVHD may manifest in the oral mucosa; however, it often develops immediately following HSCT when routine dental treatment is postponed. Chronic GVHD may manifest in the oral mucosa, the salivary glands, and the musculoskeletal compartment. It may indirectly affect the teeth and the oral flora, putting the patient at risk for infections. Importantly, GVHD poses an increased risk for oral cancer. GVHD has a wide range of oral manifestations, some of which may affect dental treatment.
Collapse
Affiliation(s)
- Michele Miranda
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Francesco Gianfreda
- Department of Industrial Engineering, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | | | - Sofia Armati
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Alberta Barlattani
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Patrizio Bollero
- Department of System Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
| |
Collapse
|
20
|
Borrill R, Poulton K, Wynn R. Immunology of cord blood T-cells favors augmented disease response during clinical pediatric stem cell transplantation for acute leukemia. Front Pediatr 2023; 11:1232281. [PMID: 37780051 PMCID: PMC10534014 DOI: 10.3389/fped.2023.1232281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/22/2023] [Indexed: 10/03/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) has been an important and efficacious treatment for acute leukemia in children for over 60 years. It works primarily through the graft-vs.-leukemia (GVL) effect, in which donor T-cells and other immune cells act to eliminate residual leukemia. Cord blood is an alternative source of stem cells for transplantation, with distinct biological and immunological characteristics. Retrospective clinical studies report superior relapse rates with cord blood transplantation (CBT), when compared to other stem cell sources, particularly for patients with high-risk leukemia. Xenograft models also support the superiority of cord blood T-cells in eradicating malignancy, when compared to those derived from peripheral blood. Conversely, CBT has historically been associated with an increased risk of transplant-related mortality (TRM) and morbidity, particularly from infection. Here we discuss clinical aspects of CBT, the unique immunology of cord blood T-cells, their role in the GVL effect and future methods to maximize their utility in cellular therapies for leukemia, honing and harnessing their antitumor properties whilst managing the risks of TRM.
Collapse
Affiliation(s)
- Roisin Borrill
- Blood and Marrow Transplant Unit, Royal Manchester Children’s Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, School of Biological Sciences, Lydia Becker Institute of Immunology and Inflammation, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kay Poulton
- Transplantation Laboratory, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Robert Wynn
- Blood and Marrow Transplant Unit, Royal Manchester Children’s Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
21
|
Buxbaum NP, Socié G, Hill GR, MacDonald KPA, Tkachev V, Teshima T, Lee SJ, Ritz J, Sarantopoulos S, Luznik L, Zeng D, Paczesny S, Martin PJ, Pavletic SZ, Schultz KR, Blazar BR. Chronic GvHD NIH Consensus Project Biology Task Force: evolving path to personalized treatment of chronic GvHD. Blood Adv 2023; 7:4886-4902. [PMID: 36322878 PMCID: PMC10463203 DOI: 10.1182/bloodadvances.2022007611] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 01/26/2023] Open
Abstract
Chronic graft-versus-host disease (cGvHD) remains a prominent barrier to allogeneic hematopoietic stem cell transplantion as the leading cause of nonrelapse mortality and significant morbidity. Tremendous progress has been achieved in both the understanding of pathophysiology and the development of new therapies for cGvHD. Although our field has historically approached treatment from an empiric position, research performed at the bedside and bench has elucidated some of the complex pathophysiology of cGvHD. From the clinical perspective, there is significant variability of disease manifestations between individual patients, pointing to diverse biological underpinnings. Capitalizing on progress made to date, the field is now focused on establishing personalized approaches to treatment. The intent of this article is to concisely review recent knowledge gained and formulate a path toward patient-specific cGvHD therapy.
Collapse
Affiliation(s)
- Nataliya P. Buxbaum
- Department of Pediatrics, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Gerard Socié
- Hematology-Transplantation, Assistance Publique-Hopitaux de Paris & University of Paris – INSERM UMR 676, Hospital Saint Louis, Paris, France
| | - Geoffrey R. Hill
- Division of Medical Oncology, The University of Washington, Seattle, WA
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Kelli P. A. MacDonald
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Victor Tkachev
- Division of Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Takanori Teshima
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Stephanie J. Lee
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Jerome Ritz
- Dana-Farber Cancer Institute, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA
| | - Stefanie Sarantopoulos
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Duke Cancer Institute, Durham, NC
| | - Leo Luznik
- Division of Hematologic Malignancies, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Defu Zeng
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute, Hematologic Maligancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA
| | - Sophie Paczesny
- Department of Microbiology and Immunology and Cancer Immunology Program, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC
| | - Paul J. Martin
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Steven Z. Pavletic
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Kirk R. Schultz
- Michael Cuccione Childhood Cancer Research Program, British Columbia Children’s Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Bruce R. Blazar
- Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, University of Minnesota, Minneappolis, MN
| |
Collapse
|
22
|
Prockop S, Wachter F. The current landscape: Allogeneic hematopoietic stem cell transplant for acute lymphoblastic leukemia. Best Pract Res Clin Haematol 2023; 36:101485. [PMID: 37611999 DOI: 10.1016/j.beha.2023.101485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 05/31/2023] [Indexed: 08/25/2023]
Abstract
One of the consistent features in development of hematopoietic stem cell transplant (HCT) for Acute Lymphoblastic Leukemia (ALL) is the rapidity with which discoveries in the laboratory are translated into innovations in clinical care. Just a few years after murine studies demonstrated that rescue from radiation induced marrow failure is mediated by cellular not humoral factors, E. Donnall Thomas reported on the transfer of bone marrow cells into irradiated leukemia patients. This was followed quickly by the first descriptions of Graft versus Leukemia (GvL) effect and Graft versus Host Disease (GvHD). Despite the pivotal nature of these findings, early human transplants were uniformly unsuccessful and identified the challenges that continue to thwart transplanters today - leukemic relapse, regimen related toxicity, and GvHD. While originally only an option for young, fit patients with a matched family donor, expansion of the donor pool to include unrelated donors, umbilical cord blood units, and more recently the growing use of haploidentical donors have all made transplant a more accessible therapy for patients with ALL. Novel agents for conditioning, prevention and treatment of GvHD have improved outcomes and investigators continue to develop novel treatment strategies that balance regimen related toxicity with disease control. Our evolving understanding of how to prevent and treat GvHD and how to prevent relapse are incorporated into novel clinical trials that are expected to further improve outcomes. Here we review current considerations and future directions for both adult and pediatric patients undergoing HCT for ALL, including indication for transplant, donor selection, cytoreductive regimens, and outcomes.
Collapse
Affiliation(s)
- Susan Prockop
- Pediatric Stem Cell Transplant Program, DFCI/BCH Center for Cancer and Blood Disorders, Pediatrics, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States.
| | - Franziska Wachter
- Pediatric Stem Cell Transplant Program, DFCI/BCH Center for Cancer and Blood Disorders, Pediatrics, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States.
| |
Collapse
|
23
|
Maurer K, Soiffer RJ. The delicate balance of graft versus leukemia and graft versus host disease after allogeneic hematopoietic stem cell transplantation. Expert Rev Hematol 2023; 16:943-962. [PMID: 37906445 PMCID: PMC11195539 DOI: 10.1080/17474086.2023.2273847] [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: 08/04/2023] [Accepted: 10/18/2023] [Indexed: 11/02/2023]
Abstract
INTRODUCTION The curative basis of allogeneic hematopoietic stem cell transplantation (HSCT) relies in part upon the graft versus leukemia (GvL) effect, whereby donor immune cells recognize and eliminate recipient malignant cells. However, alloreactivity of donor cells against recipient tissues may also be deleterious. Chronic graft versus host disease (cGvHD) is an immunologic phenomenon wherein alloreactive donor T cells aberrantly react against host tissues, leading to damaging inflammatory symptoms. AREAS COVERED Here, we discuss biological insights into GvL and cGvHD and strategies to balance the prevention of GvHD with maintenance of GvL in modern HSCT. EXPERT OPINION/COMMENTARY Relapse remains the leading cause of mortality after HSCT with rates as high as 40% for some diseases. GvHD is a major cause of morbidity after HSCT, occurring in up to half of patients and responsible for 15-20% of deaths after HSCT. Intriguingly, the development of chronic GvHD may be linked to lower relapse rates after HSCT, suggesting that GvL and GvHD may be complementary sides of the immunologic foundation of HSCT. The ability to fine tune the balance of GvL and GvHD will lead to improvements in survival, relapse rates, and quality of life for patients undergoing HSCT.
Collapse
Affiliation(s)
- Katie Maurer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Robert J Soiffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Yu T, Luo C, Zhang H, Tan Y, Yu L. Cord blood-derived CD19-specific chimeric antigen receptor T cells: an off-the-shelf promising therapeutic option for treatment of diffuse large B-cell lymphoma. Front Immunol 2023; 14:1139482. [PMID: 37449207 PMCID: PMC10338183 DOI: 10.3389/fimmu.2023.1139482] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 05/26/2023] [Indexed: 07/18/2023] Open
Abstract
Purpose Autologous chimeric antigen receptor (CAR) T cell therapy is one of the most significant breakthroughs in hematological malignancies. However, a three-week manufacturing cycle and ineffective T cell dysfunction in some patients hinder the widespread application of auto-CAR T cell therapy. Studies suggest that cord blood (CB), with its unique biological properties, could be an optimal source for CAR T cells, providing a product with 'off-the-shelf' availability. Therefore, exploring the potential of CB as an immunotherapeutic agent is essential for understanding and promoting the further use of CAR T cell therapy. Experimental design We used CB to generate CB-derived CD19-targeting CAR T (CB CD19-CAR T) cells. We assessed the anti-tumor capacity of CB CD19-CAR T cells to kill diffuse large B cell lymphoma (DLBCL) in vitro and in vivo. Results CB CD19-CAR T cells showed the target-specific killing of CD19+ T cell lymphoma cell line BV173 and CD19+ DLBCL cell line SUDHL-4, activated various effector functions, and inhibited tumor progression in a mouse (BALB/c nude) model. However, some exhaustion-associated genes were involved in off-tumor cytotoxicity towards activated lymphocytes. Gene expression profiles confirmed increased chemokines/chemokine receptors and exhaustion genes in CB CD19-CAR T cells upon tumor stimulation compared to CB T cells. They indicated inherent changes in the associated signaling pathways in the constructed CB CAR T cells and targeted tumor processes. Conclusion CB CD19-CAR T cells represent a promising therapeutic strategy for treating DLBCL. The unique biological properties and high availability of CB CD19-CAR T cells make this approach feasible.
Collapse
Affiliation(s)
- Tiantian Yu
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Division of Hematopathology and Department of Pathology, Duke University Medical Center, Durham, NC, United States
| | - Cancan Luo
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Huihui Zhang
- R&D Department, Qilu Cell Therapy Technology Co., Ltd., Jinan, Shandong, China
| | - Yi Tan
- R&D Department, Qilu Cell Therapy Technology Co., Ltd., Jinan, Shandong, China
| | - Li Yu
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
25
|
Gail LM, Schell KJ, Łacina P, Strobl J, Bolton SJ, Steinbakk Ulriksen E, Bogunia-Kubik K, Greinix H, Crossland RE, Inngjerdingen M, Stary G. Complex interactions of cellular players in chronic Graft-versus-Host Disease. Front Immunol 2023; 14:1199422. [PMID: 37435079 PMCID: PMC10332803 DOI: 10.3389/fimmu.2023.1199422] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/07/2023] [Indexed: 07/13/2023] Open
Abstract
Chronic Graft-versus-Host Disease is a life-threatening inflammatory condition that affects many patients after allogeneic hematopoietic stem cell transplantation. Although we have made substantial progress in understanding disease pathogenesis and the role of specific immune cell subsets, treatment options are still limited. To date, we lack a global understanding of the interplay between the different cellular players involved, in the affected tissues and at different stages of disease development and progression. In this review we summarize our current knowledge on pathogenic and protective mechanisms elicited by the major involved immune subsets, being T cells, B cells, NK cells and antigen presenting cells, as well as the microbiome, with a special focus on intercellular communication of these cell types via extracellular vesicles as up-and-coming fields in chronic Graft-versus-Host Disease research. Lastly, we discuss the importance of understanding systemic and local aberrant cell communication during disease for defining better biomarkers and therapeutic targets, eventually enabling the design of personalized treatment schemes.
Collapse
Affiliation(s)
- Laura Marie Gail
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Kimberly Julia Schell
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Piotr Łacina
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Johanna Strobl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Steven J. Bolton
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Katarzyna Bogunia-Kubik
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Hildegard Greinix
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Rachel Emily Crossland
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Georg Stary
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| |
Collapse
|
26
|
Malard F, Holler E, Sandmaier BM, Huang H, Mohty M. Acute graft-versus-host disease. Nat Rev Dis Primers 2023; 9:27. [PMID: 37291149 DOI: 10.1038/s41572-023-00438-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/02/2023] [Indexed: 06/10/2023]
Abstract
Acute graft-versus-host disease (GVHD) is a common immune complication that can occur after allogeneic haematopoietic cell transplantation (alloHCT). Acute GVHD is a major health problem in these patients, and is associated with high morbidity and mortality. Acute GVHD is caused by the recognition and the destruction of the recipient tissues and organs by the donor immune effector cells. This condition usually occurs within the first 3 months after alloHCT, but later onset is possible. Targeted organs include the skin, the lower and upper gastrointestinal tract and the liver. Diagnosis is mainly based on clinical examination, and complementary examinations are performed to exclude differential diagnoses. Preventive treatment for acute GVHD is administered to all patients who receive alloHCT, although it is not always effective. Steroids are used for first-line treatment, and the Janus kinase 2 (JAK2) inhibitor ruxolitinib is second-line treatment. No validated treatments are available for acute GVHD that is refractory to steroids and ruxolitinib, and therefore it remains an unmet medical need.
Collapse
Affiliation(s)
- Florent Malard
- Sorbonne Université, Centre de Recherche Saint-Antoine INSERM UMRs938, Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, AP-HP, Paris, France.
| | - Ernst Holler
- University Hospital of Regensburg, Department of Internal Medicine 3, Regensburg, Germany
| | - Brenda M Sandmaier
- Fred Hutchinson Cancer Center, Translational Science and Therapeutics Division, Seattle, WA, USA
- University of Washington School of Medicine, Division of Medical Oncology, Seattle, WA, USA
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Province, Hangzhou, China
- Engineering Laboratory for Stem Cell and Immunity Therapy, Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, China
| | - Mohamad Mohty
- Sorbonne Université, Centre de Recherche Saint-Antoine INSERM UMRs938, Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, AP-HP, Paris, France.
| |
Collapse
|
27
|
Pang Y, Holtzman NG. Immunopathogenic mechanisms and modulatory approaches to graft-versus-host disease prevention in acute myeloid leukaemia. Best Pract Res Clin Haematol 2023; 36:101475. [PMID: 37353287 PMCID: PMC10291443 DOI: 10.1016/j.beha.2023.101475] [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: 04/23/2023] [Revised: 05/03/2023] [Accepted: 05/03/2023] [Indexed: 06/25/2023]
Abstract
Allogeneic haematopoietic stem cell transplantation (HSCT) remains the only potential cure for intermediate to high-risk acute myeloid leukaemia (AML). The therapeutic effect of HSCT is largely dependent on the powerful donor-derived immune response against recipient leukaemia cells, known as graft-versus-leukaemia effect (GvL). However, the donor-derived immune system can also cause acute or chronic damage to normal recipient organs and tissues, in a process known as graft-versus-host disease (GvHD). GvHD is a leading cause of non-relapse mortality in HSCT recipients. There are many similarities and cross talk between the immune pathways of GvL and GvHD. Studies have demonstrated that both processes require the presence of mismatched alloantigens between the donor and recipient, and activation of immune responses centered around donor T-cells, which can be further modulated by various recipient or donor factors. Dissecting GvL from GvHD to achieve more effective GvHD prevention and enhanced GvL has been the holy grail of HSCT research. In this review, we focused on the key factors that contribute to the immune responses of GvL and GvHD, the effect on GvL with different GvHD prophylactic strategies, and the potential impact of various AML relapse prevention therapy or treatments on GvHD.
Collapse
Affiliation(s)
- Yifan Pang
- Department of Haematologic Oncology and Blood Disorders, Levine Cancer Institute, Charlotte, NC, USA.
| | - Noa G Holtzman
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
28
|
Patel DA, Crain M, Pusic I, Schroeder MA. Acute Graft-versus-Host Disease: An Update on New Treatment Options. Drugs 2023:10.1007/s40265-023-01889-2. [PMID: 37247105 DOI: 10.1007/s40265-023-01889-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2023] [Indexed: 05/30/2023]
Abstract
Acute graft-versus-host disease (GVHD) occurs in approximately 50% of patients and remains a primary driver of non-relapse and transplant-related mortality. The best treatment remains prevention with either in vivo or ex vivo T-cell depletion, with multiple strategies used worldwide based on factors such as institution preference, ability to perform graft manipulation, and ongoing clinical trials. Predicting patients at high risk for developing severe acute GVHD based on clinical and biomarker-based criteria allows for escalation or potential de-escalation of therapy. Modern therapies for treatment of the disease include JAK/STAT pathway inhibitors, which are standard of care in the second-line setting and are being investigated for upfront management of non-severe risk based on biomarkers. Salvage therapies beyond the second-line remain suboptimal. In this review, we will focus on the most clinically used GVHD prevention and treatment strategies, including the accumulating data on JAK inhibitors in both settings.
Collapse
Affiliation(s)
- Dilan A Patel
- Section of BMT & Leukemia, Division of Oncology, Department of Medicine, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Mallory Crain
- Section of BMT & Leukemia, Division of Oncology, Department of Medicine, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Iskra Pusic
- Section of BMT & Leukemia, Division of Oncology, Department of Medicine, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Mark A Schroeder
- Section of BMT & Leukemia, Division of Oncology, Department of Medicine, Washington University in St Louis School of Medicine, St Louis, MO, USA.
| |
Collapse
|
29
|
Mariotti J, Fowler DH, Bramanti S, Pavletic SZ. Editorial: Controversies and expectations for prevention and treatment of graft-versus-host-disease: a biological and clinical perspective. Front Immunol 2023; 14:1212756. [PMID: 37256121 PMCID: PMC10225737 DOI: 10.3389/fimmu.2023.1212756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 06/01/2023] Open
Affiliation(s)
- Jacopo Mariotti
- Bone Marrow Transplantation (BMT) and Cell Therapy Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | | | - Stefania Bramanti
- Bone Marrow Transplantation (BMT) and Cell Therapy Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Steve Z. Pavletic
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
30
|
de Witte MA, Mooyaart JE, Hoogenboom JD, Chabannon C, Malard F, Ruggeri A, Kuball J. Activity of ex vivo graft and DLI Engineering within the last decade increases, a survey from the EBMT Cellular Therapy & Immunobiology Working Party. Bone Marrow Transplant 2023:10.1038/s41409-023-01953-1. [PMID: 36934148 DOI: 10.1038/s41409-023-01953-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 02/27/2023] [Accepted: 03/08/2023] [Indexed: 03/20/2023]
Affiliation(s)
- M A de Witte
- Department of Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | - C Chabannon
- Institut Paoli-Calmettes, Centre de Lutte Contre le Cancer; Centre d'Investigations Cliniques en Biothérapies, Université d'Aix-Marseille, Inserm, CBT 1409, Marseille, France
| | - F Malard
- Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Centre de Recherche Saint-Antoine (CRSA), INSERM, Paris, France
| | - A Ruggeri
- San Raffaele Scientific Institute, Hematology and Bone marrow Transplantation Unit, Milan, Italy
| | - J Kuball
- Department of Hematology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
31
|
Fang Y, Zhu Y, Kramer A, Chen Y, Li YR, Yang L. Graft-versus-Host Disease Modulation by Innate T Cells. Int J Mol Sci 2023; 24:ijms24044084. [PMID: 36835495 PMCID: PMC9962599 DOI: 10.3390/ijms24044084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/07/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Allogeneic cell therapies, defined by genetically mismatched transplantation, have the potential to become a cost-effective solution for cell-based cancer immunotherapy. However, this type of therapy is often accompanied by the development of graft-versus-host disease (GvHD), induced by the mismatched major histocompatibility complex (MHC) between healthy donors and recipients, leading to severe complications and death. To address this issue and increase the potential for allogeneic cell therapies in clinical practice, minimizing GvHD is a crucial challenge. Innate T cells, encompassing subsets of T lymphocytes including mucosal-associated invariant T (MAIT) cells, invariant natural killer T (iNKT) cells, and gamma delta T (γδ T) cells, offer a promising solution. These cells express MHC-independent T-cell receptors (TCRs), allowing them to avoid MHC recognition and thus GvHD. This review examines the biology of these three innate T-cell populations, evaluates research on their roles in GvHD modulation and allogeneic stem cell transplantation (allo HSCT), and explores the potential futures for these therapies.
Collapse
Affiliation(s)
- Ying Fang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Yichen Zhu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Adam Kramer
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Yuning Chen
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Yan-Ruide Li
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
- Correspondence: (L.Y.); (Y.-R.L.); Tel.: +1-310-825-8609 (L.Y.); +1-310-254-6086 (Y.-R.L.)
| | - Lili Yang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
- Correspondence: (L.Y.); (Y.-R.L.); Tel.: +1-310-825-8609 (L.Y.); +1-310-254-6086 (Y.-R.L.)
| |
Collapse
|
32
|
Obermayer B, Keilholz L, Conrad T, Frentsch M, Blau IW, Vuong L, Lesch S, Movasshagi K, Tietze-Stolley C, Loyal L, Henze L, Penack O, Stervbo U, Babel N, Haas S, Beule D, Bullinger L, Wittenbecher F, Na IK. Single-cell clonal tracking of persistent T-cells in allogeneic hematopoietic stem cell transplantation. Front Immunol 2023; 14:1114368. [PMID: 36860867 PMCID: PMC9969884 DOI: 10.3389/fimmu.2023.1114368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/23/2023] [Indexed: 02/17/2023] Open
Abstract
The critical balance between intended and adverse effects in allogeneic hematopoietic stem cell transplantation (alloHSCT) depends on the fate of individual donor T-cells. To this end, we tracked αβT-cell clonotypes during stem cell mobilization treatment with granulocyte-colony stimulating factor (G-CSF) in healthy donors and for six months during immune reconstitution after transfer to transplant recipients. More than 250 αβT-cell clonotypes were tracked from donor to recipient. These clonotypes consisted almost exclusively of CD8+ effector memory T cells (CD8TEM), which exhibited a different transcriptional signature with enhanced effector and cytotoxic functions compared to other CD8TEM. Importantly, these distinct and persisting clonotypes could already be delineated in the donor. We confirmed these phenotypes on the protein level and their potential for selection from the graft. Thus, we identified a transcriptional signature associated with persistence and expansion of donor T-cell clonotypes after alloHSCT that may be exploited for personalized graft manipulation strategies in future studies.
Collapse
Affiliation(s)
- Benedikt Obermayer
- Core Unit Bioinformatics (CUBI), Berlin Institute of Health at Charite – Universitätsmedizin Berlin, Berlin, Germany
| | - Luisa Keilholz
- Department of Hematology, Oncology, and Tumor Immunology, Charite – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charite – Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas Conrad
- Core Unit Genomics, Berlin Institute of Health at Charite – Universitätsmedizin Berlin, Berlin, Germany,Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Marco Frentsch
- Department of Hematology, Oncology, and Tumor Immunology, Charite – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charite – Universitätsmedizin Berlin, Berlin, Germany
| | - Igor-Wolfgang Blau
- Department of Hematology, Oncology, and Tumor Immunology, Charite – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lam Vuong
- Department of Hematology, Oncology, and Tumor Immunology, Charite – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,Stem Cell Facility, Charite - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stella Lesch
- Department of Hematology, Oncology, and Tumor Immunology, Charite – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charite – Universitätsmedizin Berlin, Berlin, Germany
| | - Kamran Movasshagi
- Department of Hematology, Oncology, and Tumor Immunology, Charite – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,Stem Cell Facility, Charite - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Carola Tietze-Stolley
- Department of Hematology, Oncology, and Tumor Immunology, Charite – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,Stem Cell Facility, Charite - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lucie Loyal
- BIH Center for Exploratory Diagnostic Sciences (EDS), Berlin Institute of Health at Charite – Universitätsmedizin Berlin, Berlin, Germany,Si-M/”Der Simulierte Mensch” a science framework of Technische Universität Berlin and Charite - Universitätsmedizin Berlin, Berlin, Germany,Immunomics - Regenerative Immunology and Aging, Berlin Institute of Health at Charite – Universitätsmedizin Berlin, Berlin, Germany
| | - Larissa Henze
- BIH Center for Exploratory Diagnostic Sciences (EDS), Berlin Institute of Health at Charite – Universitätsmedizin Berlin, Berlin, Germany,Si-M/”Der Simulierte Mensch” a science framework of Technische Universität Berlin and Charite - Universitätsmedizin Berlin, Berlin, Germany,Immunomics - Regenerative Immunology and Aging, Berlin Institute of Health at Charite – Universitätsmedizin Berlin, Berlin, Germany
| | - Olaf Penack
- Department of Hematology, Oncology, and Tumor Immunology, Charite – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charite – Universitätsmedizin Berlin, Berlin, Germany
| | - Ulrik Stervbo
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charite – Universitätsmedizin Berlin, Berlin, Germany,Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Nina Babel
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charite – Universitätsmedizin Berlin, Berlin, Germany,Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Simon Haas
- Department of Hematology, Oncology, and Tumor Immunology, Charite – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,BIH Center for Exploratory Diagnostic Sciences (EDS), Berlin Institute of Health at Charite – Universitätsmedizin Berlin, Berlin, Germany,German Cancer Consortium (DKTK), Charite - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics (CUBI), Berlin Institute of Health at Charite – Universitätsmedizin Berlin, Berlin, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology, and Tumor Immunology, Charite – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,German Cancer Consortium (DKTK), Charite - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,ECRC Experimental and Clinical Research Center, Charite – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Friedrich Wittenbecher
- Department of Hematology, Oncology, and Tumor Immunology, Charite – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charite – Universitätsmedizin Berlin, Berlin, Germany
| | - Il-Kang Na
- Department of Hematology, Oncology, and Tumor Immunology, Charite – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charite – Universitätsmedizin Berlin, Berlin, Germany,Si-M/”Der Simulierte Mensch” a science framework of Technische Universität Berlin and Charite - Universitätsmedizin Berlin, Berlin, Germany,German Cancer Consortium (DKTK), Charite - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,ECRC Experimental and Clinical Research Center, Charite – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany,*Correspondence: Il-Kang Na,
| |
Collapse
|
33
|
Naik S, Triplett BM. Selective depletion of naïve T cells by targeting CD45RA. Front Oncol 2023; 12:1009143. [PMID: 36776371 PMCID: PMC9911795 DOI: 10.3389/fonc.2022.1009143] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/22/2022] [Indexed: 01/28/2023] Open
Affiliation(s)
- Swati Naik
- *Correspondence: Swati Naik, ; Brandon M. Triplett,
| | | |
Collapse
|
34
|
Gilman KE, Cracchiolo MJ, Matiatos AP, Davini DW, Simpson RJ, Katsanis E. Partially replacing cyclophosphamide with bendamustine in combination with cyclosporine A improves survival and reduces xenogeneic graft-versus-host-disease. Front Immunol 2023; 13:1045710. [PMID: 36700195 PMCID: PMC9868157 DOI: 10.3389/fimmu.2022.1045710] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Introduction The use of allogeneic hematopoietic cell transplantation (allo-HCT) for treating hematological disorders is increasing, but the development of graft-versus-host disease (GvHD) remains a major cause of morbidity and mortality. The use of post-transplant cyclophosphamide (CY) has significantly improved outcomes following allo-HCT, but complications of viral reactivation due to delayed immune reconstitution or relapse remain. Other laboratories are evaluating the potential benefit of lowering the dose of CY given post-transplant, whereas our laboratory has been focusing on whether partially replacing CY with another DNA alkylating agent, bendamustine (BEN) may be advantageous in improving outcomes with allo-HCT. Methods Here, we utilized a xenogeneic GvHD (xGvHD) model in which immunodeficient NSG mice are infused with human peripheral blood mononuclear cells (PBMCs). Results We show that a lower dose of CY (25 mg/kg) given on days +3 and +4 or CY (75 mg/kg) given on only day +3 post-PBMC infusion is not sufficient for improving survival from xGvHD, but can be improved with the addition of BEN (15 mg/kg) on day +4 to day +3 CY (75 mg/kg). CY/BEN treated mice when combined with cyclosporine A (CSA) (10mg/kg daily from days +5 to +18 and thrice weekly thereafter), had improved outcomes over CY/CY +CSA treated mice. Infiltration of GvHD target organs was reduced in both CY/CY and CY/BEN treatment groups versus those receiving no treatment. CY/CY +CSA mice exhibited more severe xGvHD at day 10, marked by decreased serum albumin and increased intestinal permeability. CY/BEN treated mice had reductions in naïve, effector memory and Th17 polarized T cells. RNAseq analysis of splenocytes isolated from CY/CY and CY/BEN treated animals revealed increased gene set enrichment in multiple KEGG pathways related to cell migration, proliferation/differentiation, and inflammatory pathways, among others with CY/BEN treatment. Conclusion Together, we illustrate that the use of CY/BEN is safe and shows similar control of xGvHD to CY/CY, but when combined with CSA, survival with CY/BEN is significantly prolonged compared to CY/CY.
Collapse
Affiliation(s)
- Kristy E. Gilman
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States
| | | | - Andrew P. Matiatos
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States
| | - Dan W. Davini
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States
| | - Richard J. Simpson
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States,Department of Immunobiology, University of Arizona, Tucson, AZ, United States,Department of Medicine, University of Arizona, Tucson, AZ, United States,Department of Pathology, University of Arizona, Tucson, AZ, United States
| | - Emmanuel Katsanis
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States,Department of Immunobiology, University of Arizona, Tucson, AZ, United States,School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, United States,The University of Arizona Cancer Center, Tucson, AZ, United States,Department of Pathology, University of Arizona, Tucson, AZ, United States,*Correspondence: Emmanuel Katsanis,
| |
Collapse
|
35
|
Gottlieb DJ, Sutrave G, Jiang W, Avdic S, Street JA, Simms R, Clancy LE, Antonenas V, Gloss BS, Bateman C, Bishop DC, Micklethwaite KP, Blyth E. Combining CD34+ stem cell selection with prophylactic pathogen and leukemia directed T-cell immunotherapy to simultaneously reduce graft versus host disease, infection, and leukemia recurrence after allogeneic stem cell transplant. Am J Hematol 2023; 98:159-165. [PMID: 35560045 PMCID: PMC10952473 DOI: 10.1002/ajh.26594] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 02/04/2023]
Abstract
We designed a trial to simultaneously address the problems of graft versus host disease (GVHD), infection, and recurrence of malignancy after allogeneic stem cell transplantation. CD34+ stem cell isolation was used to minimize the development of acute and chronic GVHD. Two prophylactic infusions, one combining donor-derived cytomegalovirus, Epstein-Barr virus, and Aspergillus fumigatus specific T-cells and the other comprising donor-derived CD19 directed chimeric antigen receptor (CAR) bearing T-cells, were given 21-28 days after transplant. Two patients were transplanted for acute lymphoblastic leukemia from HLA identical siblings using standard doses of cyclophosphamide and total body irradiation without antilymphocyte globulin. Patients received no post-transplant immune suppression and were given no pre-CAR T-cell lymphodepletion. Neutrophil and platelet engraftment was prompt. Following adoptive T-cell infusions, there was rapid appearance of antigen-experienced CD8+ and to a lesser extent CD4+ T-cells. Tetramer-positive T-cells targeting CMV and EBV appeared rapidly after T-cell infusion and persisted for at least 1 year. CAR T-cell expansion occurred and persisted for up to 3 months. T-cell receptor tracking confirmed the presence of product-derived T-cell clones in blood targeting all three pathogens. Both patients are alive over 3 years post-transplant without evidence of GVHD or disease recurrence. Combining robust donor T-cell depletion with directed T-cell adoptive immunotherapy targeting infectious and malignant antigens permits independent modulation of GVHD, infection, and disease recurrence. The combination may separate GVHD from the graft versus tumor effect, accelerate immune reconstitution, and improve transplant tolerability.
Collapse
Affiliation(s)
- David J. Gottlieb
- Blood Transplant and Cell Therapies ProgramWestmead HospitalSydneyNew South Wales
- Sydney Medical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South Wales
- Department of HaematologyWestmead HospitalSydneyNew South Wales
- T‐Cell Therapies GroupWestmead Institute for Medical ResearchSydneyNew South Wales
| | - Gaurav Sutrave
- Blood Transplant and Cell Therapies ProgramWestmead HospitalSydneyNew South Wales
- Sydney Medical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South Wales
- T‐Cell Therapies GroupWestmead Institute for Medical ResearchSydneyNew South Wales
| | - Wei Jiang
- Blood Transplant and Cell Therapies ProgramWestmead HospitalSydneyNew South Wales
- Sydney Medical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South Wales
| | - Selmir Avdic
- Sydney Medical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South Wales
- T‐Cell Therapies GroupWestmead Institute for Medical ResearchSydneyNew South Wales
| | - Janine A. Street
- Sydney Medical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South Wales
- T‐Cell Therapies GroupWestmead Institute for Medical ResearchSydneyNew South Wales
| | - Renee Simms
- Blood Transplant and Cell Therapies ProgramWestmead HospitalSydneyNew South Wales
- Sydney Medical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South Wales
| | - Leighton E. Clancy
- Blood Transplant and Cell Therapies ProgramWestmead HospitalSydneyNew South Wales
- T‐Cell Therapies GroupWestmead Institute for Medical ResearchSydneyNew South Wales
- Institute of Clinical Pathology and Medical ResearchNew South Wales Health PathologyWestmeadNew South WalesAustralia
| | - Vicki Antonenas
- Institute of Clinical Pathology and Medical ResearchNew South Wales Health PathologyWestmeadNew South WalesAustralia
| | - Brian S. Gloss
- Westmead Research HubWestmead Institute for Medical ResearchSydneyNew South WalesAustralia
| | - Caroline Bateman
- Sydney Medical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South Wales
- T‐Cell Therapies GroupWestmead Institute for Medical ResearchSydneyNew South Wales
- Departments of Haematology and OncologyChildren's Hospital at WestmeadSydneyNew South WalesAustralia
| | - David C. Bishop
- Blood Transplant and Cell Therapies ProgramWestmead HospitalSydneyNew South Wales
- Sydney Medical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South Wales
- T‐Cell Therapies GroupWestmead Institute for Medical ResearchSydneyNew South Wales
| | - Kenneth P. Micklethwaite
- Blood Transplant and Cell Therapies ProgramWestmead HospitalSydneyNew South Wales
- Sydney Medical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South Wales
- Department of HaematologyWestmead HospitalSydneyNew South Wales
- Institute of Clinical Pathology and Medical ResearchNew South Wales Health PathologyWestmeadNew South WalesAustralia
| | - Emily Blyth
- Blood Transplant and Cell Therapies ProgramWestmead HospitalSydneyNew South Wales
- Sydney Medical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South Wales
- Department of HaematologyWestmead HospitalSydneyNew South Wales
- T‐Cell Therapies GroupWestmead Institute for Medical ResearchSydneyNew South Wales
| |
Collapse
|
36
|
Chu Y, Talano JA, Baxter-Lowe LA, Verbsky JW, Morris E, Mahanti H, Ayello J, Keever-Taylor C, Johnson B, Weinberg RS, Shi Q, Moore TB, Fabricatore S, Grossman B, van de Ven C, Shenoy S, Cairo MS. Donor chimerism and immune reconstitution following haploidentical transplantation in sickle cell disease. Front Immunol 2022; 13:1055497. [PMID: 36569951 PMCID: PMC9780682 DOI: 10.3389/fimmu.2022.1055497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/18/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction We previously reported the initial results of a phase II multicenter transplant trial using haploidentical parental donors for children and aolescents with high-risk sickle cell disease achieving excellent survival with exceptionally low rates of graft-versus-host disease and resolution of sickle cell disease symptoms. To investigate human leukocyte antigen (HLA) sensitization, graft characteristics, donor chimerism, and immune reconstitution in these recipients. Methods CD34 cells were enriched using the CliniMACS® system with a target dose of 10 x 106 CD34+ cells/kg with a peripheral blood mononuclear cell (PBMNC) addback dose of 2x105 CD3/kg in the final product. Pre-transplant HLA antibodies were characterized. Donor chimerism was monitored 1-24 months post-transplant. Comprehensive assessment of immune reconstitution included lymphocyte subsets, plasma cytokines, complement levels, anti-viral T-cell responses, activation markers, and cytokine production. Infections were monitored. Results HLA antibodies were detected in 7 of 11 (64%) evaluable patients but rarely were against donor antigens. Myeloid engraftment was rapid (100%) at a median of 9 days. At 30 days, donor chimerism was 93-99% and natural killer cell levels were restored. By 60 days, CD19 B cells were normal. CD8 and CD4 T-cells levels were normal by 279 and 365 days, respectively. Activated CD4 and CD8 T-cells were elevated at 100-365 days post-transplant while naïve cells remained below baseline. Tregs were elevated at 100-270 days post-transplant, returning to baseline levels at one year. At one year, C3 and C4 levels were above baseline and CH50 levels were near baseline. At one year, cytokine levels were not significantly different from baseline. Discussion These results suggest that haploidentical transplantation with CD34-enriched cells and peripheral blood mononuclear cell addback results in rapid engraftment, sustained donor chimerism and broad-based immune reconstitution.
Collapse
Affiliation(s)
- Yaya Chu
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Julie-An Talano
- Department of Pediatrics, Hematology/Oncology and BMT, Children’s Hospital of Wisconsin, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Lee Ann Baxter-Lowe
- Department of Pathology, Children’s Hospital of Los Angeles, University of Southern California, Los Angeles, CA, United States
| | - James W. Verbsky
- Department of Pediatrics, Hematology/Oncology and BMT, Children’s Hospital of Wisconsin, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Erin Morris
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Harshini Mahanti
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Janet Ayello
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States,Department of Pathology, New York Medical College, Valhalla, NY, United States
| | - Carolyn Keever-Taylor
- Department of Medicine, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Bryon Johnson
- Department of Medicine, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | | | - Qiuhu Shi
- Department of Epidemiology and Community Health, New York Medical College, Valhalla, NY, United States
| | - Theodore B. Moore
- Department of Pediatrics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
| | - Sandra Fabricatore
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Brenda Grossman
- Department of Pathology and Immunology, Washington University, St Louis, MO, United States
| | - Carmella van de Ven
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Shalini Shenoy
- Department of Pediatrics and Transfusion Medicine, Washington University, St Louis, MO, United States
| | - Mitchell S. Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States,Department of Pathology, New York Medical College, Valhalla, NY, United States,Department of Medicine, New York Medical College, Valhalla, NY, United States,Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, United States,Department of Cell Biology, New York Medical College, Valhalla, NY, United States,Department of Anatomy, New York Medical College, Valhalla, NY, United States,*Correspondence: Mitchell S. Cairo,
| |
Collapse
|
37
|
Handgretinger R, Arendt AM, Maier CP, Lang P. Ex vivo and in vivo T-cell depletion in allogeneic transplantation: towards less or non-cytotoxic conditioning regimens. Expert Rev Clin Immunol 2022; 18:1285-1296. [PMID: 36220154 DOI: 10.1080/1744666x.2022.2134857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Although tremendous progress has been made since the introduction of allogeneic hematopoietic stem cell transplantation (HSCT) decades ago, there are still many obstacles to overcome. A major obstacle is the presence of T-lymphocytes in the recipient and in the donor. Recipient-derived T-lymphocytes not eliminated by the conditioning regimen are a major barrier and can lead to mixed chimerism or to complete rejection of the graft. Donor-derived T-lymphocytes can induce severe acute and chronic Graft-versus-Host Disease (GvHD). AREAS COVERED Currently published strategies for in vivo depletion of recipient-derived T-lymphocytes are discussed including the increase of the intensity of the conditioning regimen, the addition of anti-thymocyte globulin (ATG) or the anti-CD52 monoclonal antibody Campath. For the depletion or tolerization of the donor-derived T-lymphocytes, ex vivo-T-cell depletion methods, such as positive selection of CD34+ stem cells, negative depletion of CD3+ or TcRαβ+ T-lymphocytes or the use of post-transplant cyclophosphamide (PTCy) have been developed. EXPERT COMMENTARY All these currently used approaches have their disadvantages and new approaches should be investigated. In this review, we discuss current and propose new possible strategies to overcome the HLA barrier by using more specific T-cell directed therapies and/or by the combinations of current methods.
Collapse
Affiliation(s)
- Rupert Handgretinger
- Department of Hematology/Oncology. Children's University Hospital, University of Tuebingen, Germany.,Abu Dhabi Stem Cells Center, Abu Dhabi, UAE
| | - Anne-Marie Arendt
- Department of Hematology/Oncology. Children's University Hospital, University of Tuebingen, Germany
| | - Claus-Philipp Maier
- Department of Hematology/Oncology. Children's University Hospital, University of Tuebingen, Germany.,Department of Hematology, Oncology, Clinical Immunology and Rheumatology, Center for Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Peter Lang
- Department of Hematology/Oncology. Children's University Hospital, University of Tuebingen, Germany
| |
Collapse
|
38
|
Kansu E, Ward D, Sanchez AP, Cunard R, Hayran M, Huseyin B, Vaughan M, Ku G, Curtin P, Mulroney C, Costello C, Castro JE, Wieduwilt M, Corringham S, Ihasz-Davis A, Nelson C, Ball ED. Extracorporeal photopheresis for the treatment of chronic graft versus host disease. HEMATOLOGY (AMSTERDAM, NETHERLANDS) 2022; 27:785-794. [PMID: 35802815 DOI: 10.1080/16078454.2022.2095884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Chronic graft versus host disease (chronic GVHD) still remains the leading cause of late morbidity and mortality for allogeneic hematopoietic stem cell transplant (allo-HSCT) recipients. In this retrospective study, 53 consecutive allo-HSCT patients with chronic GVHD refractory to corticosteroids were treated with extracorporeal photopheresis (ECP). METHODS This study was performed as a retrospective single-center study. Medical records of a total of 59 patients treated with ECP for chronic GVHD were reviewed. RESULTS Best organ responses to ECP were observed in skin, mouth mucosa, eyes and liver. Overall response rate (ORR) to ECP was 81.2% (CR 17% and PR 64.2%). Overall survival (OS) was 84.9% and 36.7%, at 1 and 3 years, respectively. Female sex appears to have an advantage on ORR. Patients achieving ORR were able to maintain their responses with a prolonged continuation of treatments for +6 and +12 months indicating the benefits of longer ECP treatment. DISCUSSION We found that patients with chronic GVHD who were treated with ECP for 12 months or longer had a higher response rate. Our findings in line with the data reported previously suggest that patients responding to ECP should continue longer therapy schedules to achieve a better and sustained response. In our cohort, long-term ECP therapy was safe and well-tolerated with no significant adverse effects. Best responses were observed in the patients with skin, eye, liver and oral involvement. The ECP procedure offers the advantage relative to the problems with typical immunosuppressive agents. The female sex appeared to have an advantage based on the cumulative probability of the OR after ECP for chronic GVHD.
Collapse
Affiliation(s)
- Emin Kansu
- Hacettepe University Cancer Institute, Ankara, Turkey
| | - David Ward
- Division of Nephrology and Hypertension, Apheresis Unit, University of California San Diego Health, La Jolla, CA, USA
| | - Amber P Sanchez
- Division of Nephrology and Hypertension, Apheresis Unit, University of California San Diego Health, La Jolla, CA, USA
| | - Robyn Cunard
- Division of Nephrology and Hypertension, Apheresis Unit, University of California San Diego Health, La Jolla, CA, USA
| | - Mutlu Hayran
- Hacettepe University Cancer Institute, Ankara, Turkey
| | - Beril Huseyin
- Hacettepe University Cancer Institute, Ankara, Turkey
| | - Majella Vaughan
- Division of Blood and Marrow Transplantation, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, USA
| | - Grace Ku
- Genentech, Inc. South San Francisco, CA, USA
| | | | - Carolyn Mulroney
- Division of Blood and Marrow Transplantation, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, USA
| | - Caitlin Costello
- Division of Blood and Marrow Transplantation, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, USA
| | | | - Matthew Wieduwilt
- Division of Blood and Marrow Transplantation, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, USA
| | - Sue Corringham
- Division of Blood and Marrow Transplantation, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, USA
| | - Anita Ihasz-Davis
- Division of Blood and Marrow Transplantation, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, USA
| | - Connie Nelson
- Division of Blood and Marrow Transplantation, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, USA
| | - Edward D Ball
- Division of Blood and Marrow Transplantation, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, USA
| |
Collapse
|
39
|
Watkins B, Williams KM. Controversies and expectations for the prevention of GVHD: A biological and clinical perspective. Front Immunol 2022; 13:1057694. [PMID: 36505500 PMCID: PMC9726707 DOI: 10.3389/fimmu.2022.1057694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/03/2022] [Indexed: 11/24/2022] Open
Abstract
Severe acute and chronic graft versus host disease (GVHD) remains a major cause of morbidity and mortality after allogeneic hematopoietic cell transplantation. Historically, cord blood and matched sibling transplantation has been associated with the lowest rates of GVHD. Newer methods have modified the lymphocyte components to minimize alloimmunity, including: anti-thymocyte globulin, post-transplant cyclophosphamide, alpha/beta T cell depletion, and abatacept. These agents have shown promise in reducing severe GVHD, however, can be associated with increased risks of relapse, graft failure, infections, and delayed immune reconstitution. Nonetheless, these GVHD prophylaxis strategies have permitted expansion of donor sources, especially critical for those of non-Caucasian decent who previously lacked transplant options. This review will focus on the biologic mechanisms driving GVHD, the method by which each agent impacts these activated pathways, and the clinical consequences of these modern prophylaxis approaches. In addition, emerging novel targeted strategies will be described. These GVHD prophylaxis approaches have revolutionized our ability to increase access to transplant and have provided important insights into the biology of GVHD and immune reconstitution.
Collapse
Affiliation(s)
- Benjamin Watkins
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA, United States
| | | |
Collapse
|
40
|
Ibrutinib Treatment of Pediatric Chronic Graft-versus-Host Disease: Primary Results from the Phase 1/2 iMAGINE Study. Transplant Cell Ther 2022; 28:771.e1-771.e10. [DOI: 10.1016/j.jtct.2022.08.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/17/2022] [Accepted: 08/20/2022] [Indexed: 11/21/2022]
|
41
|
Hamilton BK, Cutler C, Divine C, Juckett M, LeMaistre C, Stewart S, Wilder J, Horowitz M, Khera N, Burns LJ. Are We Making PROGRESS in Preventing Graft-versus-Host Disease and Improving Clinical Outcomes? Impact of BMT CTN 1301 Study Results on Clinical Practice. Transplant Cell Ther 2022; 28:419-425. [PMID: 35550441 PMCID: PMC9364468 DOI: 10.1016/j.jtct.2022.05.002] [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: 03/04/2022] [Revised: 04/28/2022] [Accepted: 05/01/2022] [Indexed: 11/15/2022]
Abstract
The need for prospective randomized clinical trials investigating novel graft-versus-host disease (GVHD) prevention strategies that include other clinical outcomes impacted by GVHD has been highlighted as a priority for the field of hematopoietic cell transplantation. A recently completed study through the Blood and Marrow Transplant Clinical Trials Network (BMT CTN 1301) comparing CD34+ selection and post-transplantation cyclophosphamide with tacrolimus/methotrexate (Tac/MTX) for GVHD prevention demonstrated no significant differences in the primary endpoint of chronic GVHD relapse-free survival among the 3 approaches. The trial did not demonstrate a superior approach compared with Tac/MTX; however, it did highlight several challenges in determining the best and most relevant approaches to clinical trial design, particularly in the context of current and ongoing changes in real-world practices. Here we review the results of BMT CTN 1301 and their implications for clinical practice and future clinical trial design.
Collapse
Affiliation(s)
- Betty K Hamilton
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio.
| | - Corey Cutler
- Division of Stem Cell Transplantation and Cellular Therapy, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Clint Divine
- Division of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Mark Juckett
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | | | - Susan Stewart
- Blood and Marrow Transplant Information Network, Highland Park, Illinois
| | - Jennifer Wilder
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland
| | - Mary Horowitz
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Nandita Khera
- Division of Hematology and Oncology, Mayo Clinic Arizona, Phoenix, Arizona
| | - Linda J Burns
- Center for International Blood and Marrow Transplant Research, Milwaukee, Wisconsin
| |
Collapse
|
42
|
Dekker L, Sanders E, Lindemans CA, de Koning C, Nierkens S. Naive T Cells in Graft Versus Host Disease and Graft Versus Leukemia: Innocent or Guilty? Front Immunol 2022; 13:893545. [PMID: 35795679 PMCID: PMC9250980 DOI: 10.3389/fimmu.2022.893545] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
The outcome of allogeneic hematopoietic cell transplantation (allo-HCT) largely depends on the development and management of graft-versus-host disease (GvHD), infections, and the occurrence of relapse of malignancies. Recent studies showed a lower incidence of chronic GvHD and severe acute GvHD in patients receiving naive T cell depleted grafts compared to patients receiving complete T cell depleted grafts. On the other hand, the incidence of acute GvHD in patients receiving cord blood grafts containing only naive T cells is rather low, while potent graft-versus-leukemia (GvL) responses have been observed. These data suggest the significance of naive T cells as both drivers and regulators of allogeneic reactions. The naive T cell pool was previously thought to be a quiescent, homogenous pool of antigen-inexperienced cells. However, recent studies showed important differences in phenotype, differentiation status, location, and function within the naive T cell population. Therefore, the adequate recovery of these seemingly innocent T cells might be relevant in the imminent allogeneic reactions after allo-HCT. Here, an extensive review on naive T cells and their contribution to the development of GvHD and GvL responses after allo-HCT is provided. In addition, strategies specifically directed to stimulate adequate reconstitution of naive T cells while reducing the risk of GvHD are discussed. A better understanding of the relation between naive T cells and alloreactivity after allo-HCT could provide opportunities to improve GvHD prevention, while maintaining GvL effects to lower relapse risk.
Collapse
Affiliation(s)
- Linde Dekker
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Evy Sanders
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Coco de Koning
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
43
|
Song Q, Nasri U, Nakamura R, Martin PJ, Zeng D. Retention of Donor T Cells in Lymphohematopoietic Tissue and Augmentation of Tissue PD-L1 Protection for Prevention of GVHD While Preserving GVL Activity. Front Immunol 2022; 13:907673. [PMID: 35677056 PMCID: PMC9168269 DOI: 10.3389/fimmu.2022.907673] [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: 03/30/2022] [Accepted: 04/26/2022] [Indexed: 11/30/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (Allo-HCT) is a curative therapy for hematological malignancies (i.e., leukemia and lymphoma) due to the graft-versus-leukemia (GVL) activity mediated by alloreactive T cells that can eliminate residual malignant cells and prevent relapse. However, the same alloreactive T cells can cause a serious side effect, known as graft-versus-host disease (GVHD). GVHD and GVL occur in distinct organ and tissues, with GVHD occurring in target organs (e.g., the gut, liver, lung, skin, etc.) and GVL in lympho-hematopoietic tissues where hematological cancer cells primarily reside. Currently used immunosuppressive drugs for the treatment of GVHD inhibit donor T cell activation and expansion, resulting in a decrease in both GVHD and GVL activity that is associated with cancer relapse. To prevent GVHD, it is important to allow full activation and expansion of alloreactive T cells in the lympho-hematopoietic tissues, as well as prevent donor T cells from migrating into the GVHD target tissues, and tolerize infiltrating T cells via protective mechanisms, such as PD-L1 interacting with PD-1, in the target tissues. In this review, we will summarize major approaches that prevent donor T cell migration into GVHD target tissues and approaches that augment tolerization of the infiltrating T cells in the GVHD target tissues while preserving strong GVL activity in the lympho-hematopoietic tissues.
Collapse
Affiliation(s)
- Qingxiao Song
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, Unites States.,Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, Unites States.,Fujian Medical University Center of Translational Hematology, Fujian Institute of Hematology, and Fujian Medical University Union Hospital, Fuzhou, China
| | - Ubaydah Nasri
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, Unites States.,Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, Unites States
| | - Ryotaro Nakamura
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, Unites States
| | - Paul J Martin
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, United States
| | - Defu Zeng
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, Unites States.,Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, Unites States
| |
Collapse
|
44
|
Bonifacius A, Tischer-Zimmermann S, Santamorena MM, Mausberg P, Schenk J, Koch S, Barnstorf-Brandes J, Gödecke N, Martens J, Goudeva L, Verboom M, Wittig J, Maecker-Kolhoff B, Baurmann H, Clark C, Brauns O, Simon M, Lang P, Cornely OA, Hallek M, Blasczyk R, Seiferling D, Köhler P, Eiz-Vesper B. Rapid Manufacturing of Highly Cytotoxic Clinical-Grade SARS-CoV-2-specific T Cell Products Covering SARS-CoV-2 and Its Variants for Adoptive T Cell Therapy. Front Bioeng Biotechnol 2022; 10:867042. [PMID: 35480981 PMCID: PMC9036989 DOI: 10.3389/fbioe.2022.867042] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
Objectives: Evaluation of the feasibility of SARS-CoV-2-specific T cell manufacturing for adoptive T cell transfer in COVID-19 patients at risk to develop severe disease. Methods: Antiviral SARS-CoV-2-specific T cells were detected in blood of convalescent COVID-19 patients following stimulation with PepTivator SARS-CoV-2 Select using Interferon-gamma Enzyme-Linked Immunospot (IFN-γ ELISpot), SARS-CoV-2 T Cell Analysis Kit (Whole Blood) and Cytokine Secretion Assay (CSA) and were characterized with respect to memory phenotype, activation state and cytotoxic potential by multicolor flow cytometry, quantitative real-time PCR and multiplex analyses. Clinical-grade SARS-CoV-2-specific T cell products were generated by stimulation with MACS GMP PepTivator SARS-CoV-2 Select using CliniMACS Prodigy and CliniMACS Cytokine Capture System (IFN-gamma) (CCS). Functionality of enriched T cells was investigated in cytotoxicity assays and by multiplex analysis of secreted cytotoxic molecules upon target recognition. Results: Donor screening via IFN-γ ELISpot allows for pre-selection of potential donors for generation of SARS-CoV-2-specific T cells. Antiviral T cells reactive against PepTivator SARS-CoV-2 Select could be magnetically enriched from peripheral blood of convalescent COVID-19 patients by small-scale CSA resembling the clinical-grade CCS manufacturing process and showed an activated and cytotoxic T cell phenotype. Four clinical-grade SARS-CoV-2-specific T cell products were successfully generated with sufficient cell numbers and purities comparable to those observed in donor pretesting via CSA. The T cells in the generated products were shown to be capable to replicate, specifically recognize and kill target cells in vitro and secrete cytotoxic molecules upon target recognition. Cell viability, total CD3+ cell number, proliferative capacity and cytotoxic potential remained stable throughout storage of up to 72 h after end of leukapheresis. Conclusion: Clinical-grade SARS-CoV-2-specific T cells are functional, have proliferative capacity and target-specific cytotoxic potential. Their function and phenotype remain stable for several days after enrichment. The adoptive transfer of partially matched, viable human SARS-CoV-2-specific T lymphocytes collected from convalescent individuals may provide the opportunity to support the immune system of COVID-19 patients at risk for severe disease.
Collapse
Affiliation(s)
- Agnes Bonifacius
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Sabine Tischer-Zimmermann
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Maria Michela Santamorena
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Philip Mausberg
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Josephine Schenk
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Stephanie Koch
- Deutsche Gesellschaft für Gewebetransplantation, Hannover, Germany
| | - Johanna Barnstorf-Brandes
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Nina Gödecke
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Jörg Martens
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Lilia Goudeva
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Murielle Verboom
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Jana Wittig
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Britta Maecker-Kolhoff
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | | | - Caren Clark
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Olaf Brauns
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Martina Simon
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Peter Lang
- Department of Pediatric Hematology and Oncology, University Children's Hospital, University of Tuebingen, Tuebingen, Germany
| | - Oliver A Cornely
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Clinical Trials Centre Cologne (ZKS Köln), University of Cologne, Cologne, Germany.,German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Michael Hallek
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
| | - Rainer Blasczyk
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | | | - Philipp Köhler
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Britta Eiz-Vesper
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| |
Collapse
|
45
|
Soiffer RJ. Naïve T-Cell Depletion to Prevent GVHD: Searching for a Better Mousetrap. J Clin Oncol 2022; 40:1139-1141. [PMID: 35213234 DOI: 10.1200/jco.22.00105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
46
|
Takahashi T, Prockop SE. T-cell depleted haploidentical hematopoietic cell transplantation for pediatric malignancy. Front Pediatr 2022; 10:987220. [PMID: 36313879 PMCID: PMC9614427 DOI: 10.3389/fped.2022.987220] [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: 07/05/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Access to allogenic hematopoietic cell transplantation (HCT), a potentially curative treatment for chemotherapy-resistant hematologic malignancies, can be limited if no human leukocyte antigen (HLA) identical related or unrelated donor is available. Alternative donors include Cord Blood as well as HLA-mismatched unrelated or related donors. If the goal is to minimize the number of HLA disparities, partially matched unrelated donors are more likely to share 8 or 9 of 10 HLA alleles with the recipient. However, over the last decade, there has been success with haploidentical HCT performed using the stem cells from HLA half-matched related donors. As the majority of patients have at least one eligible and motivated haploidentical donor, recruitment of haploidentical related donors is frequently more rapid than of unrelated donors. This advantage in the accessibility has historically been offset by the increased risks of graft rejection, graft-versus-host disease and delayed immune reconstitution. Various ex vivo T-cell depletion (TCD) methods have been investigated to overcome the immunological barrier and facilitate immune reconstitution after a haploidentical HCT. This review summarizes historical and contemporary clinical trials of haploidentical TCD-HCT, mainly in pediatric malignancy, and describes the evolution of these approaches with a focus on serial improvements in the kinetics of immune reconstitution. Methods of TCD discussed include in vivo as well as ex vivo positive and negative selection. In addition, haploidentical TCD as a platform for post-HCT cellular therapies is discussed. The present review highlights that, as a result of the remarkable progress over half a century, haploidentical TCD-HCT can now be considered as a preferred alternative donor option for children with hematological malignancy in need of allogeneic HCT.
Collapse
Affiliation(s)
- Takuto Takahashi
- Pediatric Stem Cell Transplantation, Boston Children's Hospital/Dana-Farber Cancer Institute, Boston, MA, United States.,Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, United States
| | - Susan E Prockop
- Pediatric Stem Cell Transplantation, Boston Children's Hospital/Dana-Farber Cancer Institute, Boston, MA, United States
| |
Collapse
|