1
|
Plavsa A, Suresh T, Dalal S, Mirea L, Adams RH, Shenoy S, Ngwube A. Comparison of outcomes following subcutaneous or intravenous alemtuzumab administered prior to reduced intensity conditioning for transplantation in pediatric sickle cell disease. Transpl Immunol 2025; 89:102179. [PMID: 39904467 DOI: 10.1016/j.trim.2025.102179] [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: 06/17/2024] [Revised: 01/13/2025] [Accepted: 01/29/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Alemtuzumab-containing conditioning regimens are used for allogeneic hematopoietic stem cell transplantation (HSCT) to reduce acute and chronic graft-versus-host disease (GVHD) and the risk of graft rejection. Alemtuzumab is typically administered intravenously but is often accompanied by infusion-related side effects, including injection site reactions and anaphylaxis. Little is known about the routes of administration and if they differ in safety and efficacy in pediatric patients, especially when used in transplant conditioning. OBJECTIVES To compare adverse effects and efficacy outcomes between intravenous and subcutaneous alemtuzumab administration in pediatric patients with sickle cell disease who have undergone HSCT. STUDY DESIGN A retrospective cohort of 49 pediatric patients with sickle cell disease aged 4-16 years underwent HSCT and received either intravenous or subcutaneous alemtuzumab at St. Louis Children's Hospital or Phoenix Children's Hospital. The incidence of infusion-related reactions, neutrophil and platelet recovery, graft failure, and immune reconstitution were compared. RESULTS We found that subcutaneous alemtuzumab administration elicited fewer infusion-related reactions than intravenously administered drug (p = 0.038). No significant differences in engraftment rates, graft failure rates, infectious complications, acute GVHD, and immune reconstitution were found between the two groups. CONCLUSION Subcutaneous administration of alemtuzumab for children undergoing transplant for sickle cell disease is safe and effective.
Collapse
Affiliation(s)
- Alexandra Plavsa
- Department of Pediatrics, Creighton University School of Medicine, Phoenix, AZ, USA
| | - Tara Suresh
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Stuti Dalal
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Lucia Mirea
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ, USA
| | - Roberta H Adams
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ, USA
| | - Shalini Shenoy
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Alexander Ngwube
- Department of Pediatrics, Creighton University School of Medicine, Phoenix, AZ, USA; Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ, USA.
| |
Collapse
|
2
|
Elliott J, Koldej R, Khot A, Ritchie D. Graft-Versus-Host Disease Mouse Models: A Clinical-Translational Perspective. Methods Mol Biol 2025; 2907:1-56. [PMID: 40100591 DOI: 10.1007/978-1-0716-4430-0_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
A variety of graft-versus-host disease (GVHD) models have been developed in mice for the purpose of allowing laboratory investigation of the pathobiology, prevention, and treatment of GVHD in humans. While such models are crucial in advancing our knowledge in this field, there are some key limitations that need to be considered when translating laboratory discoveries into the clinical context. This chapter will discuss current clinical practices in transplantation and GVHD and the relative strengths and weaknesses of mouse models that attempt to replicate these states.
Collapse
Affiliation(s)
- Jessica Elliott
- ACRF Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC, Australia.
- Department of Clinical Haematology, Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- Medicine Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia.
| | - Rachel Koldej
- ACRF Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Medicine Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Amit Khot
- Department of Clinical Haematology, Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Medicine Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - David Ritchie
- ACRF Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Clinical Haematology, Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Medicine Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
3
|
Peggs KS, Albon SJ, Oporto Espuelas M, Irving C, Richardson R, Casanovas-Company J, Wallace R, Guvenel A, Ghorashian S, Collura A, Subramaniyam M, Flutter B, Popova B, Castro F, Lopes A, Champion K, Schofield O, Clifton-Hadley L, Taylor T, Farrell M, Adams S, Gilmour KC, Mackinnon S, Tholouli E, Amrolia PJ. Immunotherapy with CD25/CD71-allodepleted T cells to improve T-cell reconstitution after matched unrelated donor hematopoietic stem cell transplant: a randomized trial. Cytotherapy 2023; 25:82-93. [PMID: 36220712 DOI: 10.1016/j.jcyt.2022.08.010] [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: 04/21/2022] [Revised: 07/21/2022] [Accepted: 08/27/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND AIMS Delayed immune reconstitution is a major challenge after matched unrelated donor (MUD) stem cell transplant (SCT). In this randomized phase 2 multi-center trial, Adoptive Immunotherapy with CD25/71 allodepleted donor T cells to improve immunity after unrelated donor stem cell transplant (NCT01827579), the authors tested whether allodepleted donor T cells (ADTs) can safely be used to improve immune reconstitution after alemtuzumab-based MUD SCT for hematological malignancies. METHODS Patients received standard of care or up to three escalating doses of ADTs generated through CD25+/CD71+ immunomagnetic depletion. The primary endpoint of the study was circulating CD3+ T-cell count at 4 months post-SCT. Twenty-one patients were treated, 13 in the ADT arm and eight in the control arm. RESULTS The authors observed a trend toward improved CD3+ T-cell count at 4 months in the ADT arm versus the control arm (230/µL versus 145/µL, P = 0.18), and three ADT patients achieved normal CD3+ T-cell count at 4 months (>700/µL). The rates of significant graft-versus-host disease (GVHD) were comparable in both cohorts, with grade ≥2 acute GVHD in seven of 13 and four of eight patients and chronic GVHD in three of 13 and three of eight patients in the ADT and control arms, respectively. CONCLUSIONS These data suggest that adoptive transfer of ADTs is safe, but that in the MUD setting the benefit in terms of T-cell reconstitution is limited. This approach may be of more use in the context of more rigorous T-cell depletion.
Collapse
Affiliation(s)
- Karl S Peggs
- Department of Hematology, University College London Hospital, London, UK
| | - Sarah J Albon
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, UK; Gene and Cell Therapy, Great Ormond Street Hospital for Children, London, UK
| | - Macarena Oporto Espuelas
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, UK.
| | - Catherine Irving
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, UK; Gene and Cell Therapy, Great Ormond Street Hospital for Children, London, UK
| | - Rachel Richardson
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, UK; Gene and Cell Therapy, Great Ormond Street Hospital for Children, London, UK
| | - Joan Casanovas-Company
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, UK; Gene and Cell Therapy, Great Ormond Street Hospital for Children, London, UK
| | - Rebecca Wallace
- Gene and Cell Therapy, Great Ormond Street Hospital for Children, London, UK; Molecular Hematology Section, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Aleks Guvenel
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, UK; Gene and Cell Therapy, Great Ormond Street Hospital for Children, London, UK
| | - Sara Ghorashian
- Molecular Hematology Section, University College London Great Ormond Street Institute of Child Health, London, UK; Department of Hematology, Great Ormond Street Hospital for Children, London, UK
| | - Angela Collura
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, UK; Gene and Cell Therapy, Great Ormond Street Hospital for Children, London, UK
| | - Meera Subramaniyam
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, UK; Gene and Cell Therapy, Great Ormond Street Hospital for Children, London, UK
| | - Barry Flutter
- Gene and Cell Therapy, Great Ormond Street Hospital for Children, London, UK; Molecular Hematology Section, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Bilyana Popova
- Cancer Research UK and University College London Cancer Trials Center, London, UK
| | - Fernanda Castro
- Cancer Research UK and University College London Cancer Trials Center, London, UK
| | - Andre Lopes
- Cancer Research UK and University College London Cancer Trials Center, London, UK
| | - Kim Champion
- Cancer Research UK and University College London Cancer Trials Center, London, UK
| | - Oliver Schofield
- Cancer Research UK and University College London Cancer Trials Center, London, UK
| | - Laura Clifton-Hadley
- Cancer Research UK and University College London Cancer Trials Center, London, UK
| | - Thomas Taylor
- Department of Hematology, University College London Hospital, London, UK
| | - Maria Farrell
- Department of Hematology, Manchester Royal Infirmary, Manchester, UK
| | - Stuart Adams
- Department of Hematology, Great Ormond Street Hospital for Children, London, UK
| | - Kimberly C Gilmour
- Cell Therapy and Immunology, Camelia Botnar Laboratories, Great Ormond Street Hospital for Children, London, UK
| | - Stephen Mackinnon
- Department of Hematology, University College London Hospital, London, UK
| | - Eleni Tholouli
- Department of Hematology, Manchester Royal Infirmary, Manchester, UK
| | - Persis J Amrolia
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, UK; Department of Bone Marrow Transplantation, Great Ormond Street Hospital for Children, London, UK.
| |
Collapse
|
4
|
Haddad F, Daver N. An Update on Immune Based Therapies in Acute Myeloid Leukemia: 2021 and Beyond! ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1342:273-295. [PMID: 34972969 DOI: 10.1007/978-3-030-79308-1_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Despite advances in the treatment of acute myeloid leukemia (AML), relapse is still widely observed and represents the major cause of death among patients with AML. Treatment options in the relapse setting are limited, still relying predominantly on allogeneic hematopoietic stem cell transplantation (allo-HSCT) and cytotoxic chemotherapy, with poor outcomes. Novel targeted and venetoclax-based combinations are being investigated and have shown encouraging results. Immune checkpoint inhibitors in combination with low-intensity chemotherapy demonstrated encouraging response rates and survival among patients with relapsed and/or refractory (R/R) AML, especially in the pre- and post-allo-HSCT setting. Blocking the CD47/SIRPα pathway is another strategy that showed robust anti-leukemic activity, with a response rate of around 70% and an encouraging median overall survival in patients with newly diagnosed, higher-risk myelodysplastic syndrome and patients with AML with a TP53 mutation. One approach that was proven to be very effective in the relapsed setting of lymphoid malignancies is chimeric antigen receptor (CAR) T cells. It relies on the infusion of genetically engineered T cells capable of recognizing specific epitopes on the surface of leukemia cells. In AML, different CAR constructs with different target antigens have been evaluated and demonstrated safety and feasibility in the R/R setting. However, the difficulty of potently targeting leukemic blasts in AML while sparing normal cells represents a major limitation to their use, and strategies are being tested to overcome this obstacle. A different approach is based on endogenously redirecting the patient's system cells to target and destroy leukemic cells via bispecific T-cell engagers (BiTEs) or dual antigen receptor targeting (DARTs). Early results have demonstrated the safety and feasibility of these agents, and research is ongoing to develop BiTEs with longer half-life, allowing for less frequent administration schedules and developing them in earlier and lower disease burden settings.
Collapse
Affiliation(s)
- Fadi Haddad
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
5
|
Saad A, de Lima M, Anand S, Bhatt VR, Bookout R, Chen G, Couriel D, Di Stasi A, El-Jawahri A, Giralt S, Gutman J, Ho V, Horwitz M, Hsu J, Juckett M, Kharfan-Dabaja MA, Loren A, Meade J, Mielcarek M, Moreira J, Nakamura R, Nieto Y, Roddy J, Satyanarayana G, Schroeder M, Tan CR, Tzachanis D, Burn J, Pluchino L. Hematopoietic Cell Transplantation, Version 2.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2021; 18:599-634. [PMID: 32519831 DOI: 10.6004/jnccn.2020.0021] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Hematopoietic cell transplantation (HCT) involves the infusion of hematopoietic progenitor cells into patients with hematologic disorders with the goal of re-establishing normal hematopoietic and immune function. HCT is classified as autologous or allogeneic based on the origin of hematopoietic cells. Autologous HCT uses the patient's own cells while allogeneic HCT uses hematopoietic cells from a human leukocyte antigen-compatible donor. Allogeneic HCT is a potentially curative treatment option for patients with certain types of hematologic malignancies, and autologous HCT is primarily used to support patients undergoing high-dose chemotherapy. Advances in HCT methods and supportive care in recent decades have led to improved survival after HCT; however, disease relapse and posttransplant complications still commonly occur in both autologous and allogeneic HCT recipients. Allogeneic HCT recipients may also develop acute and/or chronic graft-versus-host disease (GVHD), which results in immune-mediated cellular injury of several organs. The NCCN Guidelines for Hematopoietic Cell Transplantation focus on recommendations for pretransplant recipient evaluation and the management of GVHD in adult patients with malignant disease.
Collapse
Affiliation(s)
- Ayman Saad
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Marcos de Lima
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | | | | | | | | | | | | | | | - Vincent Ho
- Dana-Farber/Brigham and Women's Cancer Center
| | | | | | | | | | - Alison Loren
- Abramson Cancer Center at the University of Pennsylvania
| | - Javier Meade
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | - Marco Mielcarek
- Fred Hutchinson Cancer Research Center/ Seattle Cancer Care Alliance
| | - Jonathan Moreira
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | - Yago Nieto
- The University of Texas MD Anderson Cancer Center
| | - Juliana Roddy
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | | | - Mark Schroeder
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | | |
Collapse
|
6
|
Shaw BE, Jimenez-Jimenez AM, Burns LJ, Logan BR, Khimani F, Shaffer BC, Shah NN, Mussetter A, Tang XY, McCarty JM, Alavi A, Farhadfar N, Jamieson K, Hardy NM, Choe H, Ambinder RF, Anasetti C, Perales MA, Spellman SR, Howard A, Komanduri KV, Luznik L, Norkin M, Pidala JA, Ratanatharathorn V, Confer DL, Devine SM, Horowitz MM, Bolaños-Meade J. National Marrow Donor Program-Sponsored Multicenter, Phase II Trial of HLA-Mismatched Unrelated Donor Bone Marrow Transplantation Using Post-Transplant Cyclophosphamide. J Clin Oncol 2021; 39:1971-1982. [PMID: 33905264 PMCID: PMC8260905 DOI: 10.1200/jco.20.03502] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/01/2021] [Accepted: 03/16/2021] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Hematopoietic cell transplantation (HCT) is curative for hematologic disorders, but outcomes are historically inferior when using HLA-mismatched donors. Despite unrelated donor registries listing > 38 million volunteers, 25%-80% of US patients lack an HLA-matched unrelated donor, with significant disparity across ethnic groups. We hypothesized that HCT with a mismatched unrelated donor (MMUD) using post-transplant cyclophosphamide (PTCy), a novel strategy successful in overcoming genetic disparity using mismatched related donors, would be feasible and increase access to HCT. PATIENTS AND METHODS We performed a prospective phase II study of MMUD bone marrow HCT with PTCy for patients with hematologic malignancies. The primary end point was 1-year overall survival (OS), hypothesized to be 65% or better. 80 patients enrolled at 11 US transplant centers (December 2016-March 2019). Following myeloablative or reduced-intensity conditioning-based HCT, patients received PTCy on days +3, +4, with sirolimus and mycophenolate mofetil starting on day +5. We compared outcomes to Center for International Blood and Marrow Transplant Research contemporary controls receiving PTCy. RESULTS Notably, 48% of patients enrolled were ethnic minorities. 39% of pairs were matched for 4-6 out of 8 HLA alleles. The primary end point was met, with 1-year OS of 76% (90% CI, 67.3 to 83.3) in the entire cohort, and 72% and 79% in the myeloablative and reduced-intensity conditioning strata, respectively. Secondary end points related to engraftment and graft-versus-host-disease were reached. Multivariate analysis comparing the study group with other mismatched HCT controls found no significant differences in OS. CONCLUSION Our prospective study demonstrates the feasibility and effectiveness of HCT with an MMUD in the setting of PTCy. Remarkably, nearly half of the study participants belonged to an ethnic minority population, suggesting this approach may significantly expand access to HCT.
Collapse
Affiliation(s)
- Bronwen E. Shaw
- Center for International Blood and Marrow Transplant Research/Medical College of Wisconsin, Milwaukee, WI
| | | | - Linda J. Burns
- Center for International Blood and Marrow Transplant Research/Medical College of Wisconsin, Milwaukee, WI
| | - Brent R. Logan
- Center for International Blood and Marrow Transplant Research/Medical College of Wisconsin, Milwaukee, WI
| | - Farhad Khimani
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Brian C. Shaffer
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nirav N. Shah
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Alisha Mussetter
- Center for International Blood and Marrow Transplant Research/National Marrow Donor Program, Minneapolis, MN
| | - Xiao-Ying Tang
- Center for International Blood and Marrow Transplant Research/Medical College of Wisconsin, Milwaukee, WI
| | - John M. McCarty
- Massey Cancer Center Bone Marrow Transplant Program, Virginia Commonwealth University, Richmond, VA
| | - Asif Alavi
- Department of Oncology, Blood and Marrow Stem Cell Transplant Program, Karmanos Cancer Institute/Wayne State University, Detroit, MI
| | - Nosha Farhadfar
- Division of Hematology/Oncology, University of Florida College of Medicine, Gainesville, FL
| | | | - Nancy M. Hardy
- Greenebaum Cancer Center, University of Maryland, Baltimore, MD
| | - Hannah Choe
- James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Richard F. Ambinder
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | | | - Miguel-Angel Perales
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Stephen R. Spellman
- Center for International Blood and Marrow Transplant Research/National Marrow Donor Program, Minneapolis, MN
| | - Alan Howard
- Center for International Blood and Marrow Transplant Research/National Marrow Donor Program, Minneapolis, MN
| | | | - Leo Luznik
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Maxim Norkin
- LifeSouth Community Blood Centers, Baptist MD Anderson Cancer Center, Jacksonville, FL
| | | | - Voravit Ratanatharathorn
- Department of Oncology, Blood and Marrow Stem Cell Transplant Program, Karmanos Cancer Institute/Wayne State University, Detroit, MI
| | - Dennis L. Confer
- Center for International Blood and Marrow Transplant Research/National Marrow Donor Program, Minneapolis, MN
| | - Steven M. Devine
- Center for International Blood and Marrow Transplant Research/National Marrow Donor Program, Minneapolis, MN
| | - Mary M. Horowitz
- Center for International Blood and Marrow Transplant Research/Medical College of Wisconsin, Milwaukee, WI
| | | |
Collapse
|
7
|
Acharya UH, Walter RB. Chimeric Antigen Receptor (CAR)-Modified Immune Effector Cell Therapy for Acute Myeloid Leukemia (AML). Cancers (Basel) 2020; 12:E3617. [PMID: 33287224 PMCID: PMC7761730 DOI: 10.3390/cancers12123617] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/28/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
Despite the availability of an increasing number of targeted therapeutics and wider use of allogeneic hematopoietic stem cell transplantation, many patients with acute myeloid leukemia (AML) ultimately succumb to this disease. Given their remarkable efficacy in B-acute lymphoblastic leukemia and other CD19-expressing B cell malignancies, there is hope adoptive cellular transfer, particularly chimeric antigen receptor (CAR)-modified immune effector cell (IEC) therapies, may afford a novel, potent immune-based approach for the treatment of AML that complements or replaces existing ones and improves cure rates. However, it is unclear how best to translate the success of these therapies from B cell malignancies, where use of highly potent immunotherapies is facilitated by identified target antigens with near ubiquitous expression on malignant cells and non-fatal consequences from "on-target, off-tumor cell" toxicities. Herein, we review the current status of CAR-modified IEC therapies for AML, with considerations regarding suitable, relatively leukemia-restricted target antigens, expected toxicities, and interactions of the engineered cells with a profoundly immunosuppressive tumor microenvironment that restricts their therapeutic efficacy. With these challenges in mind, we will discuss possible strategies to improve the cells' potency as well as their therapeutic window for optimal clinical use in AML.
Collapse
Affiliation(s)
- Utkarsh H. Acharya
- Divisions of Hematologic Malignancies & Immune Effector Cell Therapy, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Roland B. Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA;
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA 98195, USA
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA 98195, USA
- Department of Epidemiology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
8
|
Lin WY, Wang HH, Chen YW, Lin CF, Fan HC, Lee YY. Gene Modified CAR-T Cellular Therapy for Hematologic Malignancies. Int J Mol Sci 2020; 21:ijms21228655. [PMID: 33212810 PMCID: PMC7697548 DOI: 10.3390/ijms21228655] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/15/2020] [Accepted: 11/15/2020] [Indexed: 02/06/2023] Open
Abstract
With advances in the understanding of characteristics of molecules, specific antigens on the surface of hematological malignant cells were identified and multiple therapies targeting these antigens as neoplasm treatments were developed. Among them, chimeric antigen receptor (CAR) T-cell therapy, which got United States Food and Drug Administration (FDA) approval for relapsed/refractory (r/r) diffuse large B-cell lymphoma (DLBCL) as well as for recurrent acute lymphoblastic leukemia (ALL) within the past five years, and for r/r mantle cell lymphoma (MCL) this year, represents one of the most rapidly evolving immunotherapies. Nevertheless, its applicability to other hematological malignancies, as well as its efficacy and persistence are fraught with clinical challenges. Currently, more than one thousand clinical trials in CAR T-cell therapy are ongoing and its development is changing rapidly. This review introduces the current status of CAR T-cell therapy in terms of the basic molecular aspects of CAR T-cell therapy, its application in hematological malignancies, adverse reactions during clinical use, remaining challenges, and future utilization.
Collapse
Affiliation(s)
- Wen-Ying Lin
- Department of Internal Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
| | - Hsin-Hui Wang
- Department of Pediatrics, Division of Pediatric Immunology and Nephrology, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
- Department of Pediatrics, Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan
| | - Yi-Wei Chen
- Division of Radiation Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
| | - Chun-Fu Lin
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Hueng-Chuen Fan
- Department of Pediatrics, Tungs’ Taichung Metroharbor Hospital, Wuchi, Taichung 435403, Taiwan;
- Department of Medical Research, Tungs’ Taichung Metroharbor Hospital, Wuchi, Taichung 435403, Taiwan
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan
- Department of Rehabilitation, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 356, Taiwan
| | - Yi-Yen Lee
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Correspondence: ; Tel.: +886-2-28757491; Fax: +886-2-28757588
| |
Collapse
|
9
|
Vandenhove B, Canti L, Schoemans H, Beguin Y, Baron F, Graux C, Kerre T, Servais S. How to Make an Immune System and a Foreign Host Quickly Cohabit in Peace? The Challenge of Acute Graft- Versus-Host Disease Prevention After Allogeneic Hematopoietic Cell Transplantation. Front Immunol 2020; 11:583564. [PMID: 33193397 PMCID: PMC7609863 DOI: 10.3389/fimmu.2020.583564] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/21/2020] [Indexed: 01/16/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation (alloHCT) has been used as cellular immunotherapy against hematological cancers for more than six decades. Its therapeutic efficacy relies on the cytoreductive effects of the conditioning regimen but also on potent graft-versus-tumor (GVT) reactions mediated by donor-derived immune cells. However, beneficial GVT effects may be counterbalanced by acute GVHD (aGVHD), a systemic syndrome in which donor immune cells attack healthy tissues of the recipient, resulting in severe inflammatory lesions mainly of the skin, gut, and liver. Despite standard prophylaxis regimens, aGVHD still occurs in approximately 20–50% of alloHCT recipients and remains a leading cause of transplant-related mortality. Over the past two decades, advances in the understanding its pathophysiology have helped to redefine aGVHD reactions and clinical presentations as well as developing novel strategies to optimize its prevention. In this review, we provide a brief overview of current knowledge on aGVHD immunopathology and discuss current approaches and novel strategies being developed and evaluated in clinical trials for aGVHD prevention. Optimal prophylaxis of aGVHD would prevent the development of clinically significant aGVHD, while preserving sufficient immune responsiveness to maintain beneficial GVT effects and immune defenses against pathogens.
Collapse
Affiliation(s)
- Benoît Vandenhove
- Laboratory of Hematology, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium
| | - Lorenzo Canti
- Laboratory of Hematology, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium
| | - Hélène Schoemans
- Department of Clinical Hematology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Yves Beguin
- Laboratory of Hematology, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium.,Department of Clinical Hematology, CHU of Liège, University of Liège, Liège, Belgium
| | - Frédéric Baron
- Laboratory of Hematology, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium.,Department of Clinical Hematology, CHU of Liège, University of Liège, Liège, Belgium
| | - Carlos Graux
- Department of Clinical Hematology, CHU UCL Namur (Godinne), Université Catholique de Louvain, Yvoir, Belgium
| | - Tessa Kerre
- Hematology Department, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Sophie Servais
- Laboratory of Hematology, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium.,Department of Clinical Hematology, CHU of Liège, University of Liège, Liège, Belgium
| |
Collapse
|
10
|
Mardiana S, Gill S. CAR T Cells for Acute Myeloid Leukemia: State of the Art and Future Directions. Front Oncol 2020; 10:697. [PMID: 32435621 PMCID: PMC7218049 DOI: 10.3389/fonc.2020.00697] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/14/2020] [Indexed: 12/27/2022] Open
Abstract
Relapse after conventional chemotherapy remains a major problem in patients with myeloid malignancies such as acute myeloid leukemia (AML), and the major cause of death after diagnosis of AML is from relapsed disease. The only potentially curative treatment option currently available is allogeneic hematopoietic stem cell transplantation (allo-HSCT), which through its graft-vs.-leukemia effects has the ability to eliminate residual leukemia cells. Despite its long history of success however, relapse following allo-HSCT is still a major challenge and is associated with poor prognosis. In the field of adoptive therapy, CD19-targeted chimeric antigen receptor (CAR) T cells have yielded remarkable clinical success in certain types of B-cell malignancies, and substantial efforts aimed at translating this success to myeloid malignancies are currently underway. While complete ablation of CD19-expressing B cells, both cancerous and healthy, is clinically tolerated, the primary challenge limiting the use of CAR T cells in myeloid malignancies is the absence of a dispensable antigen, as myeloid antigens are often co-expressed on normal hematopoietic stem/progenitor cells (HSPCs), depletion of which would lead to intolerable myeloablation. This review provides a discussion on the current state of CAR T cell therapy in myeloid malignancies, limitations for clinical translation, as well as the most recent approaches to overcome these barriers, through various genetic modification and combinatorial strategies in an attempt to make CAR T cell therapy a safe and viable option for patients with myeloid malignancies.
Collapse
Affiliation(s)
- Sherly Mardiana
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
- Division of Hematology-Oncology and Center for Cellular Immunotherapies, University of Pennsylvania, PA, United States
| | - Saar Gill
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
- Division of Hematology-Oncology and Center for Cellular Immunotherapies, University of Pennsylvania, PA, United States
| |
Collapse
|
11
|
Hashmi H, Bhandari S, Dhanoa J, Wu X, Rai S, Figg L, Baize T, Krem M, Hegazi M, Emmons R. Clinical Outcomes Using Mycophenolate and Tacrolimus for Graft-versus-Host Disease Prophylaxis in Patients Undergoing Allogeneic Stem Cell Transplant: A Single Institution Experience. Cureus 2020; 12:e6893. [PMID: 32190456 PMCID: PMC7058400 DOI: 10.7759/cureus.6893] [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] [Indexed: 11/05/2022] Open
Abstract
For recipients of allogeneic hematopoietic stem cell transplant (HSCT), mycophenolate mofetil (MMF) plus tacrolimus combination is mostly used in reduced-intensity (RIC), and nonmyeloablative conditioning (NMAC) whereas methotrexate and tacrolimus combination is preferred in myeloablative conditioning (MAC). We present single institution outcomes in patients undergoing allogeneic HSCT with both MAC and NMAC/RIC regimen using MMF and tacrolimus for graft-versus-host disease (GVHD) prophylaxis. Data from all adult patients who underwent allogeneic HSCT from 2007 to 2017 was collected from Data Back to Centers web-based application of Center for International Blood and Marrow Transplant Research (CIBMTR). A total of 150 patients were included with the mean age of 46.9 years. For the patients who received MAC (n=109), the cumulative incidence of grade II-IV acute GVHD at day 100 was 37%, grade II-IV acute GVHD at one year was 51%, and chronic GVHD at one year was 38%. For the patients who received NMAC/RIC (n=41), the cumulative incidence of grade II-IV acute GVHD at day 100 was 31%, grade II-IV acute GVHD at one year was 28%, and chronic GVHD at one year was 36%. This institutional analysis shows that the combination of MMF and tacrolimus yields acceptable outcomes for the prevention of acute and chronic GVHD.
Collapse
Affiliation(s)
- Hamza Hashmi
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer and Research Center, Tampa, USA
| | - Shruti Bhandari
- Division of Medical Oncology and Hematology, University of Louisville, Louisville, USA
| | - Jugraj Dhanoa
- Internal Medicine, University of Louisville, School of Medicine, Louisville, USA
| | - Xiaoyong Wu
- Bioinformatics and Biostatistics, University of Louisville, School of Public Health and Information Science, Louisville, USA
| | - Shesh Rai
- Bioinformatics and Biostatistics, University of Louisville, School of Public Health and Information Science, Louisville, USA
| | - Lindsay Figg
- Division of Clinical Oncology Pharmacy, University of Louisville Hospital, Louisville, USA
| | - Timothy Baize
- Division of Clinical Oncology Pharmacy, University of Louisville Hospital, Louisville, USA
| | - Maxwell Krem
- Division of Hematology and Blood and Marrow Transplant, University of Kentucky, Lexington, USA
| | - Mohamed Hegazi
- Division of Blood and Marrow Transplant, University of Louisville, Louisville, USA
| | - Robert Emmons
- Division of Blood and Marrow Transplant, University of Louisville, Louisville, USA
| |
Collapse
|
12
|
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-SCT) is the most established and commonly used cellular immunotherapy in cancer care. It is the most potent anti-leukemic therapy in patients with acute myeloid leukemia (AML) and is routinely used with curative intent in patients with intermediate and poor risk disease. Donor T cells, and possibly other immune cells, eliminate residual leukemia cells after prior (radio)chemotherapy. This immune-mediated response is known as graft-versus-leukemia (GvL). Donor alloimmune responses can also be directed against healthy tissues, which is known as graft-versus-host disease (GvHD). GvHD and GvL often co-occur and, therefore, a major barrier to exploiting the full immunotherapeutic benefit of donor immune cells against patient leukemia is the immunosuppression required to treat GvHD. However, curative responses to allo-SCT and GvHD do not always occur together, suggesting that these two immune responses could be de-coupled in some patients. To make further progress in successfully promoting GvL without GvHD, we must transform our limited understanding of the cellular and molecular basis of GvL and GvHD. Specifically, in most patients we do not understand the antigenic basis of immune responses in GvL and GvHD. Identification of antigens important for GvL but not GvHD, and vice versa, could impact on donor selection, allow us to track GvL immune responses and begin to specifically harness and strengthen anti-leukemic immune responses against patient AML cells, whilst minimizing the toxicity of GvHD.
Collapse
Affiliation(s)
- Connor Sweeney
- MRC Molecular Haematology Unit, Oxford Biomedical Research Centre, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Department of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Paresh Vyas
- MRC Molecular Haematology Unit, Oxford Biomedical Research Centre, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Department of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
13
|
Asai J, Yamaguchi J, Tsukamoto T, Chinen Y, Shimura Y, Kobayashi T, Katoh N, Kuroda J. Treatment rationale and design of a phase II study of narrow-band ultraviolet B phototherapy for cutaneous steroid-refractory acute graft-vs-host disease following allogenic stem-cell transplantation. Medicine (Baltimore) 2019; 98:e16372. [PMID: 31305435 PMCID: PMC6641799 DOI: 10.1097/md.0000000000016372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Acute graft-vs-host disease (aGVHD) is a common complication of allogenic hematopoietic stem-cell transplantation (allo-HSCT) and skin is the most common and often the 1st site at which aGVHD develops. Cutaneous aGVHD is usually treated with oral and/or topical corticosteroids as the 1st-line treatment; however, steroid-refractory aGVHD not only impairs patients' quality of life but also causes significant morbidity and mortality after allo-HSCT. Narrow-band ultraviolet B (NB-UVB) phototherapy has been utilized for a wide range of immunologic inflammatory skin diseases, but there is limited information on the efficacy, safety, and biomarkers for response prediction of NB-UVB for cutaneous aGVHD. AIMS The purpose of this study is to investigate the efficacy and safety of NB-UVB phototherapy for steroid-refractory cutaneous aGVHD. PATIENTS AND METHODS A total of 40 subjects aged from 16 to 70 years with steroid-refractory cutaneous aGVHD after allo-HSCT will be included in the trial. Patients with worse than stage 2 intestine/liver aGVHD will be excluded. Eligible patients will undergo NB-UVB phototherapy until resolution or further worsening of rash or occurrence of an unmanageable adverse event. The primary endpoint is the overall response rate. The secondary outcomes include rates for complete response, partial response, stable disease, progressive disease, duration of response, sparing effect on calcineurin inhibitors and/or corticosteroids, safety, and predictive biomarkers for treatment response. ETHICS AND DISSEMINATION The protocol has been approved by the institutional Clinical Research Review Board of Kyoto Prefectural University of Medicine. Written informed consent will be obtained from all patients before registration, in accordance with the Declaration of Helsinki. Results of the study will be disseminated via publications in peer-reviewed journals. TRIAL REGISTRATION Trial registration numbers UMIN000032426 and jRCTs052180005.
Collapse
Affiliation(s)
| | - Junko Yamaguchi
- Division of Hematology and Oncology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Taku Tsukamoto
- Division of Hematology and Oncology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Yoshiaki Chinen
- Division of Hematology and Oncology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Yuji Shimura
- Division of Hematology and Oncology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Tsutomu Kobayashi
- Division of Hematology and Oncology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | | | - Junya Kuroda
- Division of Hematology and Oncology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| |
Collapse
|
14
|
Poonsombudlert K, Kewcharoen J, Kanitsoraphan C, Prueksapraopong C, Limpruttidham N. Chronic graft-versus-host disease and the risk of primary disease relapse: A meta-analysis. Exp Hematol 2019; 74:33-41. [DOI: 10.1016/j.exphem.2019.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/28/2019] [Accepted: 04/23/2019] [Indexed: 01/22/2023]
|
15
|
van der Maas NG, Berghuis D, van der Burg M, Lankester AC. B Cell Reconstitution and Influencing Factors After Hematopoietic Stem Cell Transplantation in Children. Front Immunol 2019; 10:782. [PMID: 31031769 PMCID: PMC6473193 DOI: 10.3389/fimmu.2019.00782] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/25/2019] [Indexed: 12/20/2022] Open
Abstract
B cell reconstitution after hematopoietic stem cell transplantation (HSCT) is variable and influenced by different patient, donor, and treatment related factors. In this review we describe B cell reconstitution after pediatric allogeneic HST, including the kinetics of reconstitution of the different B cell subsets and the development of the B cell repertoire, and discuss the influencing factors. Observational studies show important roles for stem cell source, conditioning regimen, and graft vs. host disease in B cell reconstitution. In addition, B cell recovery can play an important role in post-transplant infections and vaccine responses to encapsulated bacteria, such as pneumococcus. A substantial number of patients experience impaired B cell function and/or dependency on Ig substitution after allogeneic HSCT. The underlying mechanisms are largely unresolved. The integrated aspects of B cell recovery after HSCT, especially BCR repertoire reconstitution, are awaiting further investigation using modern techniques in order to gain more insight into B cell reconstitution and to develop strategies to improve humoral immunity after allogeneic HSCT.
Collapse
Affiliation(s)
- Nicolaas G van der Maas
- Willem-Alexander Children's Hospital, Department of Pediatrics and Laboratory for Pediatric Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Dagmar Berghuis
- Willem-Alexander Children's Hospital, Department of Pediatrics and Laboratory for Pediatric Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Mirjam van der Burg
- Willem-Alexander Children's Hospital, Department of Pediatrics and Laboratory for Pediatric Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Arjan C Lankester
- Willem-Alexander Children's Hospital, Department of Pediatrics and Laboratory for Pediatric Immunology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
16
|
Shaw P, Shizuru J, Hoenig M, Veys P. Conditioning Perspectives for Primary Immunodeficiency Stem Cell Transplants. Front Pediatr 2019; 7:434. [PMID: 31781522 PMCID: PMC6851055 DOI: 10.3389/fped.2019.00434] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/07/2019] [Indexed: 01/10/2023] Open
Abstract
The majority of children undergoing Hematopoietic Stem cell Transplantation (HSCT) require conditioning therapy to make space and prevent rejection of the donor stem cells. The exception is certain children with Severe Combined immune deficiency, who have limited or no ability to reject the donor graft. Transplant conditioning is associated with significant morbidity and mortality from both direct toxic effects of chemotherapy as well as opportunistic infections associated with profound immunosuppression. The ultimate goal of transplant practice is to achieve sufficient engraftment of donor cells to correct the underlying disease with minimal short- and long-term toxicity to the recipient. Traditional combinations, such as busulfan and cyclophosphamide, achieve a high rate of full donor engraftment, but are associated with significant acute transplant-related-mortality and late effects such as infertility. Less "intensive" approaches, such as combinations of treosulfan or melphalan with fludarabine, are less toxic, but may be associated with rejection or low level chimerism requiring the need for re-transplantation. The major benefit of these novel approaches, however, which we hope will be realized in the decades to come, may be the preservation of fertility. Future approaches look to replace chemotherapy with non-toxic antibody conditioning. The lessons learnt in refining conditioning for HSCT are likely to be equally applicable to gene therapy protocols for the same diseases.
Collapse
Affiliation(s)
- Peter Shaw
- Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Judith Shizuru
- Departments of Medicine and Pediatrics, Stanford University, Stanford, CA, United States
| | - Manfred Hoenig
- Klinik für Kinder- und Jugendmedizin, Universitätsklinikum Ulm, Ulm, Germany
| | - Paul Veys
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | | |
Collapse
|
17
|
Villa NY, McFadden G. Virotherapy as Potential Adjunct Therapy for Graft-Vs-Host Disease. CURRENT PATHOBIOLOGY REPORTS 2018; 6:247-263. [PMID: 30595970 PMCID: PMC6290699 DOI: 10.1007/s40139-018-0186-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW This review discusses the pathophysiology, risk factors, and the advances in the prevention or treatment of graft-vs-host disease (GvHD) by exploiting adjunct virotherapy. In addition, nonviral adjunct therapeutic options for the prevention of GvHD in the context of allogeneic hematopoietic stem cell transplantation (allo-HSCT) are discussed. The role of oncolytic viruses to treat different HSCT-eligible hematological cancers is also considered and correlated with the issue of GvHD in the context of allo-HSCT. RECENT FINDINGS Emerging therapies focused on the prevention or treatment of GvHD include the use of regulatory T cells (Tregs), mesenchymal stem cells (MSCs), microbiome manipulation, B cell inhibitors, among others. Our lab and others have reported that an oncolytic DNA virus from the Poxviridae family, called myxoma virus (MYXV), not only exhibits oncolytic activity against various hematologic malignancies like multiple myeloma (MM) or acute myeloid leukemia (AML) but also, in addition, ex vivo MYXV treatment of human allogeneic-bone marrow transplants (allo-BMT), or allo-peripheral blood mononuclear cell (allo-PBMC) transplants can abrogate GvHD in xenografted mice without impairing graft-vs-tumor (GvT) effects against residual cancer. To date, this is the first and the only oncolytic virus with a dual potential of mediating oncolysis against a residual cancer target and also inhibiting or preventing GvHD following allo-HSCT. SUMMARY This review discusses how oncolytic virotherapy can be applied as a potential adjunct therapy for the potential treatment of GvHD. In addition, we highlight major emerging nonviral therapies currently studied for the treatment or prevention of GvHD. We also review the emerging oncolytic virotherapies against different hematological cancers currently eligible for allo-HSCT and highlight the potential role of the oncolytic virus MYXV to decrease GvHD while maintaining or enhancing the positive benefits of GvT.
Collapse
Affiliation(s)
- Nancy Y. Villa
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ 85287 USA
| | - Grant McFadden
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ 85287 USA
| |
Collapse
|
18
|
The case for plerixafor to replace filgrastim as the optimal agent to mobilize peripheral blood donors for allogeneic hematopoietic cell transplantation. Exp Hematol 2018; 70:1-9. [PMID: 30428338 DOI: 10.1016/j.exphem.2018.11.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/03/2018] [Accepted: 11/06/2018] [Indexed: 12/25/2022]
Abstract
Granulocyte colony-stimulating factor (G-CSF)-stimulated peripheral blood progenitor cells (G-PBs) from either a related or unrelated donor continue to be the preferred donor source for most allogeneic hematopoietic cell transplantation (HCT). Recently, the American Society for Blood and Marrow Transplantation has recommended marrow instead of G-PBs as an unrelated graft source due to its lower rate of chronic graft-versus-host disease (cGVHD). However, the use of marrow is limited by both clinical considerations (slower rate of engraftment and increased donor morbidity) and logistical considerations (use of operating room resources and increased physician utilization), so this recommendation has not been widely adopted. An optimal donor source would include the rapid engraftment characteristic and the low donor morbidity associated with G-PBs and a rate of cGVHD similar to or lower than that of marrow. Recent data suggest that plerixafor mobilized PBs (P-PBs) have the rapid engraftment characteristics of G-PBs in allogeneic HCT with less cGVHD. The biologic mechanism of the lower rate of cGVHD appears to be through mobilization of regulator natural killer cells and plasmacytoid dendritic cell precursors that are associated with lower acute and chronic GVHD compared with G-PBs and rapid engraftment characterized by rapid myeloid-repopulating capacity. We suggest that, based on the experience of the two Phase II clinical trials and the unique biology of plerixafor-mobilized donor product, it should be evaluated in Phase III trials as an approach to replacing G-CSF mobilization for allogeneic HCT.
Collapse
|