1
|
Cimons JM, DeGolier KR, Burciaga SD, Yarnell MC, Novak AJ, Rivera-Reyes AM, Kohler ME, Fry TJ. T-bet overexpression enhances CAR T cell effector functions and antigen sensitivity. J Immunother Cancer 2025; 13:e010962. [PMID: 40246581 PMCID: PMC12007057 DOI: 10.1136/jitc-2024-010962] [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: 11/01/2024] [Accepted: 04/07/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND T cells modified to express a chimeric antigen receptor (CAR) are successful against B-lineage malignancies but fail to induce durable remissions in up to half of patients and have shown limited efficacy against other types of cancer. Strategies to improve CAR T cell potency and responses to low antigen densities without inducing CAR T cell dysfunction or limiting persistence are necessary to expand durability of remissions. METHODS We overexpressed T-bet in human and mouse CAR T cells to mimic exposure to signal 3 cytokines during T cell priming to promote T helper cell 1 (Th1) polarization of CD4+CAR T cells with the goal of enhancing antitumor activity. Using human CAR T cells and xenograft models we interrogated the impact of T-bet overexpression on CAR T cell antitumor activity in vitro and in vivo. We also used a syngeneic murine CAR T cell model to study the impact of T-bet overexpression on long-term persistence and secondary responses to tumor rechallenge. RESULTS T-bet overexpression reduced expression of the Th2 cytokine interleukin 4 and promoted polyfunctional production of Th1-associated cytokines in response to CAR stimulation. T-bet overexpression enhanced some effector functions in vitro but did not improve CAR T cell-mediated control of leukemia expressing high levels of antigen in vivo. T-bet overexpression also improved effector function of murine CD19 CAR T cells with no impairment to the persistence or ability of persistent CAR T cells to re-expand and clear a secondary leukemia challenge. Finally, T-bet overexpression promoted enhanced in vitro function against leukemia expressing low levels of CD19, which translated to improved control of CD19lo leukemia in vivo by human C19 CAR T cells containing a 4-1BB costimulatory domain. CONCLUSIONS Together, our data demonstrate that T-bet overexpression induces a reduction in Th2 cytokine production, an increase in polyfunctional Th1 cytokine production and enhances 4-1BB CAR T cell activity against cancers expressing low levels of target antigen without promoting a loss in functional CAR T cell persistence.
Collapse
Affiliation(s)
- Jennifer M Cimons
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Kole R DeGolier
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Samuel D Burciaga
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Michael C Yarnell
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Amanda J Novak
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Amalia M Rivera-Reyes
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - M Eric Kohler
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
- Pediatrics, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Terry J Fry
- University of Colorado Denver Children's Hospital Colorado Research Institute, Aurora, Colorado, USA
| |
Collapse
|
2
|
Kondo T, Bourassa FXP, Achar S, DuSold J, Céspedes PF, Ando M, Dwivedi A, Moraly J, Chien C, Majdoul S, Kenet AL, Wahlsten M, Kvalvaag A, Jenkins E, Kim SP, Ade CM, Yu Z, Gaud G, Davila M, Love P, Yang JC, Dustin ML, Altan-Bonnet G, François P, Taylor N. Engineering TCR-controlled fuzzy logic into CAR T cells enhances therapeutic specificity. Cell 2025:S0092-8674(25)00290-9. [PMID: 40220754 DOI: 10.1016/j.cell.2025.03.017] [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: 01/12/2024] [Revised: 09/16/2024] [Accepted: 03/09/2025] [Indexed: 04/14/2025]
Abstract
Chimeric antigen receptor (CAR) T cell immunotherapy represents a breakthrough in the treatment of hematological malignancies, but poor specificity has limited its applicability to solid tumors. By contrast, natural T cells harboring T cell receptors (TCRs) can discriminate between neoantigen-expressing cancer cells and self-antigen-expressing healthy tissues but have limited potency against tumors. We used a high-throughput platform to systematically evaluate the impact of co-expressing a TCR and CAR on the same CAR T cell. While strong TCR-antigen interactions enhanced CAR activation, weak TCR-antigen interactions actively antagonized their activation. Mathematical modeling captured this TCR-CAR crosstalk in CAR T cells, allowing us to engineer dual TCR/CAR T cells targeting neoantigens (HHATL8F/p53R175H) and human epithelial growth factor receptor 2 (HER2) ligands, respectively. These T cells exhibited superior anti-cancer activity and minimal toxicity against healthy tissue compared with conventional CAR T cells in a humanized solid tumor mouse model. Harnessing pre-existing inhibitory crosstalk between receptors, therefore, paves the way for the design of more precise cancer immunotherapies.
Collapse
Affiliation(s)
- Taisuke Kondo
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - François X P Bourassa
- Department of Physics, McGill University, Montréal, QC, Canada; Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC, Canada
| | - Sooraj Achar
- Immunodynamics Group, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA; Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Justyn DuSold
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Pablo F Céspedes
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK; CAMS Oxford Institute, University of Oxford, Oxford, UK
| | - Makoto Ando
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Alka Dwivedi
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Josquin Moraly
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Christopher Chien
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Saliha Majdoul
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Adam L Kenet
- Immunodynamics Group, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Madison Wahlsten
- Immunodynamics Group, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Audun Kvalvaag
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK; Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Oslo, Norway
| | - Edward Jenkins
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Sanghyun P Kim
- Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Catherine M Ade
- Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Zhiya Yu
- Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Guillaume Gaud
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Marco Davila
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Paul Love
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - James C Yang
- Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Grégoire Altan-Bonnet
- Immunodynamics Group, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | - Paul François
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC, Canada; MILA Québec, Montréal, QC, Canada.
| | - Naomi Taylor
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA; Université de Montpellier, Institut de Génétique Moléculaire de Montpellier, Montpellier, France.
| |
Collapse
|
3
|
DeGolier KR, Danis E, D'Antonio M, Cimons J, Yarnell M, Kedl RM, Kohler ME, Scott-Browne JP, Fry TJ. Antigen experience history directs distinct functional states of CD8 + CAR T cells during the antileukemia response. Nat Immunol 2025; 26:68-81. [PMID: 39747430 PMCID: PMC11695263 DOI: 10.1038/s41590-024-02034-1] [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: 12/05/2023] [Accepted: 11/11/2024] [Indexed: 01/04/2025]
Abstract
Although chimeric antigen receptor (CAR) T cells are effective against B-lineage malignancies, post-CAR relapse is common, and efficacy in other tumors is limited. These challenges may be addressed through rational manipulations to control CAR T cell function. Here we examine the impact of cognate T cell antigen experience on subsequent CD8+ CAR T cell activity. Prior antigen encounter resulted in superior effector function against leukemia expressing low target antigen density at the expense of reduced proliferative capacity and susceptibility to dysfunction at limiting CAR doses. Distinctive temporal transcriptomic and epigenetic profiles in naive-derived and memory-derived CAR T cells identified RUNX family transcription factors as potential targets to augment the function of naive-derived CD8+ CAR T cells. RUNX2 overexpression enhanced antitumor efficacy of mouse CAR T cells, dependent on prior cell state, and heightened human CAR T cell functions. Our data demonstrate that prior antigen experience of CAR T cells determines functional attributes and amenability to transcription factor-mediated functional enhancement.
Collapse
Affiliation(s)
- Kole R DeGolier
- Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Etienne Danis
- Biostatistics and Bioinformatics Shared Resource, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Marc D'Antonio
- Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jennifer Cimons
- Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael Yarnell
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA
| | - Ross M Kedl
- Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - M Eric Kohler
- Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA
| | - James P Scott-Browne
- Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, USA
| | - Terry J Fry
- Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA.
| |
Collapse
|
4
|
Yao Y, Zhou J, Li Y, Shi S, Yu L, Wu D, Wang Y. CD19 CAR T-cell therapy in relapsed TCF3-HLF-positive B-cell acute lymphoblastic leukemia. Ann Hematol 2024:10.1007/s00277-024-05945-z. [PMID: 39145779 DOI: 10.1007/s00277-024-05945-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 08/12/2024] [Indexed: 08/16/2024]
Affiliation(s)
- Yao Yao
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jin Zhou
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yanting Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Sensen Shi
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Lei Yu
- Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd, Shanghai, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| | - Ying Wang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| |
Collapse
|
5
|
Wu MH, Valenca-Pereira F, Cendali F, Giddings EL, Pham-Danis C, Yarnell MC, Novak AJ, Brunetti TM, Thompson SB, Henao-Mejia J, Flavell RA, D'Alessandro A, Kohler ME, Rincon M. Deleting the mitochondrial respiration negative regulator MCJ enhances the efficacy of CD8 + T cell adoptive therapies in pre-clinical studies. Nat Commun 2024; 15:4444. [PMID: 38789421 PMCID: PMC11126743 DOI: 10.1038/s41467-024-48653-y] [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: 10/17/2023] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
Mitochondrial respiration is essential for the survival and function of T cells used in adoptive cellular therapies. However, strategies that specifically enhance mitochondrial respiration to promote T cell function remain limited. Here, we investigate methylation-controlled J protein (MCJ), an endogenous negative regulator of mitochondrial complex I expressed in CD8 cells, as a target for improving the efficacy of adoptive T cell therapies. We demonstrate that MCJ inhibits mitochondrial respiration in murine CD8+ CAR-T cells and that deletion of MCJ increases their in vitro and in vivo efficacy against murine B cell leukaemia. Similarly, MCJ deletion in ovalbumin (OVA)-specific CD8+ T cells also increases their efficacy against established OVA-expressing melanoma tumors in vivo. Furthermore, we show for the first time that MCJ is expressed in human CD8 cells and that the level of MCJ expression correlates with the functional activity of CD8+ CAR-T cells. Silencing MCJ expression in human CD8 CAR-T cells increases their mitochondrial metabolism and enhances their anti-tumor activity. Thus, targeting MCJ may represent a potential therapeutic strategy to increase mitochondrial metabolism and improve the efficacy of adoptive T cell therapies.
Collapse
Affiliation(s)
- Meng-Han Wu
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Felipe Valenca-Pereira
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Francesca Cendali
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Emily L Giddings
- Division of Immunobiology, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Catherine Pham-Danis
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplant, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Michael C Yarnell
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplant, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Amanda J Novak
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplant, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Tonya M Brunetti
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Scott B Thompson
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Jorge Henao-Mejia
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard A Flavell
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - M Eric Kohler
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplant, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA.
| | - Mercedes Rincon
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.
- Division of Immunobiology, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA.
| |
Collapse
|
6
|
Hay ZL, Kim DD, Cimons JM, Knapp JR, Kohler ME, Quansah M, Zúñiga TM, Camp FA, Fujita M, Wang XJ, O’Connor BP, Slansky JE. Granzyme F: Exhaustion Marker and Modulator of Chimeric Antigen Receptor T Cell-Mediated Cytotoxicity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1381-1391. [PMID: 38416029 PMCID: PMC10984789 DOI: 10.4049/jimmunol.2300334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 01/03/2024] [Indexed: 02/29/2024]
Abstract
Granzymes are a family of proteases used by CD8 T cells to mediate cytotoxicity and other less-defined activities. The substrate and mechanism of action of many granzymes are unknown, although they diverge among the family members. In this study, we show that mouse CD8+ tumor-infiltrating lymphocytes (TILs) express a unique array of granzymes relative to CD8 T cells outside the tumor microenvironment in multiple tumor models. Granzyme F was one of the most highly upregulated genes in TILs and was exclusively detected in PD1/TIM3 double-positive CD8 TILs. To determine the function of granzyme F and to improve the cytotoxic response to leukemia, we constructed chimeric Ag receptor T cells to overexpress a single granzyme, granzyme F or the better-characterized granzyme A or B. Using these doubly recombinant T cells, we demonstrated that granzyme F expression improved T cell-mediated cytotoxicity against target leukemia cells and induced a form of cell death other than chimeric Ag receptor T cells expressing only endogenous granzymes or exogenous granzyme A or B. However, increasing expression of granzyme F also had a detrimental impact on the viability of the host T cells, decreasing their persistence in circulation in vivo. These results suggest a unique role for granzyme F as a marker of terminally differentiated CD8 T cells with increased cytotoxicity, but also increased self-directed cytotoxicity, suggesting a potential mechanism for the end of the terminal exhaustion pathway.
Collapse
Affiliation(s)
- Zachary L.Z. Hay
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Dale D. Kim
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jennifer M. Cimons
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jennifer R. Knapp
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, 80206, USA
| | - M. Eric Kohler
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
- Center for Cancer and Blood Disorders, Children’s Hospital Colorado and Department of Pediatrics, Aurora, CO, USA
| | - Mary Quansah
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Tiffany M. Zúñiga
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Faye A. Camp
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Mayumi Fujita
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA and Department of Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, CO 80045, USA
| | - Xiao-Jing Wang
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA and Department of Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, CO 80045, USA
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO, USA, and since moved to Department of Pathology and Laboratory Medicine, University of California Davis, CA, USA
| | - Brian P. O’Connor
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, 80206, USA
| | - Jill E. Slansky
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
7
|
Knight E T, Oluwole O, Kitko C. The Implementation of Chimeric Antigen Receptor (CAR) T-cell Therapy in Pediatric Patients: Where Did We Come From, Where Are We Now, and Where are We Going? Clin Hematol Int 2024; 6:96-115. [PMID: 38817691 PMCID: PMC11108586 DOI: 10.46989/001c.94386] [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: 01/17/2024] [Accepted: 02/13/2024] [Indexed: 06/01/2024] Open
Abstract
CD19-directed Chimeric Antigen Receptor (CAR) T-cell therapy has revolutionized the treatment of patients with B-cell acute lymphoblastic leukemia (B-ALL). Somewhat uniquely among oncologic clinical trials, early clinical development occurred simultaneously in both children and adults. In subsequent years however, the larger number of adult patients with relapsed/refractory (r/r) malignancies has led to accelerated development of multiple CAR T-cell products that target a variety of malignancies, resulting in six currently FDA-approved for adult patients. By comparison, only a single CAR-T cell therapy is approved by the FDA for pediatric patients: tisagenlecleucel, which is approved for patients ≤ 25 years with refractory B-cell precursor ALL, or B-cell ALL in second or later relapse. Tisagenlecleucel is also under evaluation in pediatric patients with relapsed/refractory B-cell non-Hodgkin lymphoma, but is not yet been approved for this indication. All the other FDA-approved CD19-directed CAR-T cell therapies available for adult patients (axicabtagene ciloleucel, brexucabtagene autoleucel, and lisocabtagene maraleucel) are currently under investigations among children, with preliminary results available in some cases. As the volume and complexity of data continue to grow, so too does the necessity of rapid assimilation and implementation of those data. This is particularly true when considering "atypical" situations, e.g. those arising when patients do not precisely conform to the profile of those included in pivotal clinical trials, or when alternative treatment options (e.g. hematopoietic stem cell transplantation (HSCT) or bispecific T-cell engagers (BITEs)) are also available. We have therefore developed a relevant summary of the currently available literature pertaining to the use of CD19-directed CAR-T cell therapies in pediatric patients, and sought to provide guidance for clinicians seeking additional data about specific clinical situations.
Collapse
Affiliation(s)
| | - Olalekan Oluwole
- Medicine Hematology and Oncology, Vanderbilt University Medical Center
| | | |
Collapse
|
8
|
Wang L, Leach V, Muthusamy N, Byrd J, Long M. A CD3 humanized mouse model unmasked unique features of T-cell responses to bispecific antibody treatment. Blood Adv 2024; 8:470-481. [PMID: 37871327 PMCID: PMC10837186 DOI: 10.1182/bloodadvances.2023010971] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/04/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023] Open
Abstract
ABSTRACT T-cell bispecific antibodies (T-BsAbs) such as blinatumomab hold great promise for cancer immunotherapy. A better understanding of the in vivo immune response induced by T-BsAbs is crucial to improving their efficacy and safety profile. However, such efforts are hindered by the limitations of current preclinical models. To address this, we developed a syngeneic murine model with humanized CD3 and target antigen (CD20). This model enables the development of disseminated leukemia with a high tumor burden, which mirrors clinical findings in human patients with relapsed/refractory acute lymphoblastic leukemia. Treatment of this model with T-BsAbs results in cytokine release syndrome, with cytokine profiles and levels reflecting observations made in human patients. This model also faithfully recapitulates the dynamics of T-cell activation seen in human patients, including the temporary disappearance of T cells from the bloodstream. During this phase, T cells are sequestered in secondary lymphoid organs and undergo activation. Clinical correlative studies that rely primarily on peripheral blood samples are likely to overlook this critical activation stage, leading to a substantial underestimation of the extent of T-cell activation. Furthermore, we demonstrate that surface expression of the T-BsAb target antigen by leukemia cells triggers a swift immune response, promoting their own rejection. Humanizing the target antigen in the recipient mice is crucial to facilitate tolerance induction and successful establishment of high tumor burden. Our findings underscore the importance of meticulously optimized syngeneic murine models for investigating T-BsAb-induced immune responses and for translational research aimed at improving efficacy and safety.
Collapse
Affiliation(s)
- Lingling Wang
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Vincent Leach
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Natarajan Muthusamy
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - John Byrd
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Meixiao Long
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| |
Collapse
|
9
|
DeGolier KR, Danis E, D'Antonio M, Cimons J, Yarnell M, Kedl RM, Kohler ME, Scott-Browne JP, Fry TJ. Antigen experience history directs distinct functional states of CD8+ CAR T cells during the anti-leukemia response. RESEARCH SQUARE 2023:rs.3.rs-3712137. [PMID: 38196657 PMCID: PMC10775394 DOI: 10.21203/rs.3.rs-3712137/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Chimeric antigen receptor T cells are an effective therapy for B-lineage malignancies. However, many patients relapse and this therapeutic has yet to show strong efficacy in other hematologic or solid tumors. One opportunity for improvement lies in the ability to generate T cells with desirable functional characteristics. Here, we dissect the biology of CD8+ CAR T cells (CAR8) by controlling whether the T cell has encountered cognate TCR antigen prior to CAR generation. We find that prior antigen experience influences multiple aspects of in vitro and in vivo CAR8 functionality, resulting in superior effector function and leukemia clearance in the setting of limiting target antigen density compared to antigen-inexperienced T cells. However, this comes at the expense of inferior proliferative capacity, susceptibility to phenotypic exhaustion and dysfunction, and inability to clear wildtype leukemia in the setting of limiting CAR+ cell dose. Epigenomic and transcriptomic comparisons of these cell populations identified overexpression of the Runx2 transcription factor as a novel strategy to enhance CAR8 function, with a differential impact depending on prior cell state. Collectively, our data demonstrate that prior antigen experience determines functional attributes of a CAR T cell, as well as amenability to functional enhancement by transcription factor modulation.
Collapse
Affiliation(s)
- Kole R DeGolier
- Department of Immunology, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
| | - Etienne Danis
- Biostatistics and Bioinformatics Shared Resource, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
| | - Marc D'Antonio
- Department of Immunology, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
| | - Jennifer Cimons
- Department of Immunology, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
| | - Michael Yarnell
- Department of Pediatrics, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
- Center for Cancer and Blood Disorders, Children's Hospital Colorado; Aurora, CO, USA
| | - Ross M Kedl
- Department of Immunology, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
| | - M Eric Kohler
- Department of Immunology, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
- Center for Cancer and Blood Disorders, Children's Hospital Colorado; Aurora, CO, USA
| | - James P Scott-Browne
- Department of Immunology, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, USA
| | - Terry J Fry
- Department of Immunology, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
- Center for Cancer and Blood Disorders, Children's Hospital Colorado; Aurora, CO, USA
| |
Collapse
|
10
|
Patterson MT, Khan SM, Nunes NS, Fletcher RE, Bian J, Hadjis AD, Eckhaus MA, Mendu SK, de Paula Pohl A, Venzon DJ, Choo-Wosoba H, Ishii K, Qin H, Fry TJ, Cam M, Kanakry CG. Murine allogeneic CAR T cells integrated before or early after posttransplant cyclophosphamide exert antitumor effects. Blood 2023; 141:659-672. [PMID: 36201744 PMCID: PMC9979711 DOI: 10.1182/blood.2022016660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/20/2022] Open
Abstract
Relapse limits the therapeutic efficacy both of chimeric antigen receptor (CAR) T cells and allogeneic hematopoietic cell transplantation (allo-HCT). Patients may undergo these therapies sequentially to prevent or treat relapsed malignancy. However, direct integration of the 2 therapies has been avoided over concerns for potential induction of graft-versus-host disease (GVHD) by allogeneic CAR T cells. We have shown in murine T-cell-replete MHC-haploidentical allo-HCT that suppressive mechanisms induced immediately after posttransplant cyclophosphamide (PTCy), given on days +3/+4, prevent GVHD induction by alloreactive T cells infused as early as day +5. Therefore, we hypothesized that allogeneic CAR T cells given in a similarly integrated manner in our murine MHC-haploidentical allo-HCT model may safely exert antitumor effects. Indeed, allogeneic anti-CD19 CAR T cells given early after (day +5) PTCy or even prior to (day 0) PTCy cleared leukemia without exacerbating the cytokine release syndrome occurring from the MHC-haploidentical allo-HCT or interfering with PTCy-mediated GVHD prevention. Meanwhile, CAR T-cell treatment on day +9 or day +14 was safe but less effective, suggesting a limited therapeutic window. CAR T cells infused before PTCy were not eliminated, but surviving CAR T cells continued to proliferate highly and expand despite PTCy. In comparison with infusion on day +5, CAR T-cell infusion on day 0 demonstrated superior clinical efficacy associated with earlier CAR T-cell expansion, higher phenotypic CAR T-cell activation, less CD4+CD25+Foxp3+ CAR T-cell recovery, and transcriptional changes suggesting increased activation of CD4+ CAR T cells and more cytotoxic CD8+ CAR T cells. This study provides mechanistic insight into PTCy's impact on graft-versus-tumor immunity and describes novel approaches to integrate CAR T cells and allo-HCT that may compensate for deficiencies of each individual approach.
Collapse
Affiliation(s)
- Michael T. Patterson
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Shanzay M. Khan
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Natalia S. Nunes
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Rochelle E. Fletcher
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jing Bian
- Collaborative Bioinformatics Resource, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ashley D. Hadjis
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Michael A. Eckhaus
- Division of Veterinary Resources, Office of Research Services, National Institutes of Health, Bethesda, MD
| | - Suresh K. Mendu
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Alessandra de Paula Pohl
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - David J. Venzon
- Biostatistics and Data Management Section, Office of the Clinical Director, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Hyoyoung Choo-Wosoba
- Biostatistics and Data Management Section, Office of the Clinical Director, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Kazusa Ishii
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Haiying Qin
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Terry J. Fry
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Maggie Cam
- Collaborative Bioinformatics Resource, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Christopher G. Kanakry
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
11
|
Zebley CC, Youngblood B. Mechanisms of T cell exhaustion guiding next-generation immunotherapy. Trends Cancer 2022; 8:726-734. [PMID: 35570136 PMCID: PMC9388609 DOI: 10.1016/j.trecan.2022.04.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/08/2022] [Accepted: 04/14/2022] [Indexed: 10/18/2022]
Abstract
The functional decline in T cells during their chronic stimulation, commonly referred to as T cell exhaustion, is a major limitation for current immunotherapy approaches. As modern medicine embraces therapeutic approaches that exploit the immuno-oncology interface, a primary question is how is T cell function maintained over time in scenarios of prolonged tumor burden. Deciphering the molecular mechanisms of T cell exhaustion is now enabling the field to begin using cardinal features of T cell differentiation to develop biomarkers that can delineate responders from nonresponders prior to treatment with T cell-based therapeutics. Furthermore, applying principles of basic T cell immunity toward the development of cancer treatments is laying a foundation for rational approaches to improve immunotherapy by redirecting T cells away from a dysfunctional developmental trajectory.
Collapse
Affiliation(s)
- Caitlin C Zebley
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Ben Youngblood
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
12
|
Tracy SI, Venkatesh H, Hekim C, Heltemes-Harris LM, Knutson TP, Bachanova V, Farrar MA. Combining nilotinib and PD-L1 blockade reverses CD4+ T-cell dysfunction and prevents relapse in acute B-cell leukemia. Blood 2022; 140:335-348. [PMID: 35275990 PMCID: PMC9335501 DOI: 10.1182/blood.2021015341] [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: 12/31/2021] [Accepted: 02/28/2022] [Indexed: 11/20/2022] Open
Abstract
Patients with acute lymphoblastic leukemia have experienced significantly improved outcomes due to the advent of chimeric antigen receptor (CAR) T cells and bispecific T-cell engagers, although a proportion of patients still relapse despite these advances. T-cell exhaustion has been recently suggested to be an important driver of relapse in these patients. Indeed, phenotypic exhaustion of CD4+ T cells is predictive of relapse and poor overall survival in B-cell acute lymphoblastic leukemia (B-ALL). Thus, therapies that counter T-cell exhaustion, such as immune checkpoint blockade, may improve leukemia immunosurveillance and prevent relapse. Here, we used a murine model of Ph+ B-ALL as well as human bone marrow biopsy samples to assess the fundamental nature of CD4+ T-cell exhaustion and the preclinical therapeutic potential for combining anti-PD-L1 based checkpoint blockade with tyrosine kinase inhibitors targeting the BCR-ABL oncoprotein. Single-cell RNA-sequence analysis revealed that B-ALL induces a unique subset of CD4+ T cells with both cytotoxic and helper functions. Combination treatment with the tyrosine kinase inhibitor nilotinib and anti-PD-L1 dramatically improves long-term survival of leukemic mice. Depletion of CD4+ T cells prior to therapy completely abrogates the survival benefit, implicating CD4+ T cells as key drivers of the protective anti-leukemia immune response. Indeed, treatment with anti-PD-L1 leads to clonal expansion of leukemia-specific CD4+ T cells with the aforementioned helper/cytotoxic phenotype as well as reduced expression of exhaustion markers. These findings support efforts to use PD1/PD-L1 checkpoint blockade in clinical trials and highlight the importance of CD4+ T-cell dysfunction in limiting the endogenous anti-leukemia response.
Collapse
Affiliation(s)
- Sean I Tracy
- Center for Immunology
- Masonic Cancer Center
- Division of Hematology, Oncology and Transplantation, Department of Medicine, and
| | - Hrishi Venkatesh
- Center for Immunology
- Masonic Cancer Center
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Can Hekim
- Center for Immunology
- Masonic Cancer Center
- Orion Corporation, R&D, Immuno-Oncology Unit Tengströminkatu, Turku, Finland; and
| | - Lynn M Heltemes-Harris
- Center for Immunology
- Masonic Cancer Center
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Todd P Knutson
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN
| | - Veronika Bachanova
- Masonic Cancer Center
- Division of Hematology, Oncology and Transplantation, Department of Medicine, and
| | - Michael A Farrar
- Center for Immunology
- Masonic Cancer Center
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
13
|
B-cell acute lymphoblastic leukemia promotes an immune suppressive microenvironment that can be overcome by IL-12. Sci Rep 2022; 12:11870. [PMID: 35831470 PMCID: PMC9279427 DOI: 10.1038/s41598-022-16152-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/05/2022] [Indexed: 12/14/2022] Open
Abstract
Immunotherapies have revolutionized the treatment of B-cell acute lymphoblastic leukemia (B-ALL), but the duration of responses is still sub-optimal. We sought to identify mechanisms of immune suppression in B-ALL and strategies to overcome them. Plasma collected from children with B-ALL with measurable residual disease after induction chemotherapy showed differential cytokine expression, particularly IL-7, while single-cell RNA-sequencing revealed the expression of genes associated with immune exhaustion in immune cell subsets. We also found that the supernatant of leukemia cells suppressed T-cell function ex vivo. Modeling B-ALL in mice, we observed an altered tumor immune microenvironment, including compromised activation of T-cells and dendritic cells (DC). However, recombinant IL-12 (rIL-12) treatment of mice with B-ALL restored the levels of several pro-inflammatory cytokines and chemokines in the bone marrow and increased the number of splenic and bone marrow resident T-cells and DCs. RNA-sequencing of T-cells isolated from vehicle and rIL-12 treated mice with B-ALL revealed that the leukemia-induced increase in genes associated with exhaustion, including Lag3, Tigit, and Il10, was abrogated with rIL-12 treatment. In addition, the cytolytic capacity of T-cells co-cultured with B-ALL cells was enhanced when IL-12 and blinatumomab treatments were combined. Overall, these results demonstrate that the leukemia immune suppressive microenvironment can be restored with rIL-12 treatment which has direct therapeutic implications.
Collapse
|
14
|
Saad AA. Targeting cancer-associated glycans as a therapeutic strategy in leukemia. ALL LIFE 2022. [DOI: 10.1080/26895293.2022.2049901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Affiliation(s)
- Ashraf Abdullah Saad
- Unit of Pediatric Hematologic Oncology and BMT, Sultan Qaboos University Hospital, Muscat, Oman
| |
Collapse
|
15
|
Simonetta F, Lohmeyer JK, Hirai T, Maas-Bauer K, Alvarez M, Wenokur AS, Baker J, Aalipour A, Ji X, Haile S, Mackall CL, Negrin RS. Allogeneic CAR Invariant Natural Killer T Cells Exert Potent Antitumor Effects through Host CD8 T-Cell Cross-Priming. Clin Cancer Res 2021; 27:6054-6064. [PMID: 34376537 PMCID: PMC8563377 DOI: 10.1158/1078-0432.ccr-21-1329] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/13/2021] [Accepted: 07/30/2021] [Indexed: 01/09/2023]
Abstract
PURPOSE The development of allogeneic chimeric antigen receptor (CAR) T-cell therapies for off-the-shelf use is a major goal that faces two main immunologic challenges, namely the risk of graft-versus-host disease (GvHD) induction by the transferred cells and the rejection by the host immune system limiting their persistence. In this work we assessed the direct and indirect antitumor effect of allogeneic CAR-engineered invariant natural killer T (iNKT) cells, a cell population without GvHD-induction potential that displays immunomodulatory properties. EXPERIMENTAL DESIGN After assessing murine CAR iNKT cells direct antitumor effects in vitro and in vivo, we employed an immunocompetent mouse model of B-cell lymphoma to assess the interaction between allogeneic CAR iNKT cells and endogenous immune cells. RESULTS We demonstrate that allogeneic CAR iNKT cells exerted potent direct and indirect antitumor activity when administered across major MHC barriers by inducing tumor-specific antitumor immunity through host CD8 T-cell cross-priming. CONCLUSIONS In addition to their known direct cytotoxic effect, allogeneic CAR iNKT cells induce host CD8 T-cell antitumor responses, resulting in a potent antitumor effect lasting longer than the physical persistence of the allogeneic cells. The utilization of off-the-shelf allogeneic CAR iNKT cells could meet significant unmet needs in the clinic.
Collapse
Affiliation(s)
- Federico Simonetta
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California
- Division of Hematology, Department of Oncology, Geneva University Hospitals and Translational Research Centre in Onco-Haematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Juliane K Lohmeyer
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Toshihito Hirai
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Kristina Maas-Bauer
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Maite Alvarez
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Arielle S Wenokur
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Jeanette Baker
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Amin Aalipour
- Department of Bioengineering, Stanford University School of Medicine, Stanford, California
- Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Xuhuai Ji
- Human Immune Monitoring Center, Stanford University School of Medicine, Stanford, California
| | - Samuel Haile
- Department of Pediatrics, Stanford University, Stanford, California
| | - Crystal L Mackall
- Department of Pediatrics, Stanford University, Stanford, California
- Stanford Cancer Institute, Stanford University, Stanford, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Robert S Negrin
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
16
|
Liao W, Kohler ME, Fry T, Ernst P. Does lineage plasticity enable escape from CAR-T cell therapy? Lessons from MLL-r leukemia. Exp Hematol 2021; 100:1-11. [PMID: 34298117 PMCID: PMC8611617 DOI: 10.1016/j.exphem.2021.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 01/20/2023]
Abstract
The clinical success of engineered, CD19-directed chimeric antigen receptor (CAR) T cells in relapsed, refractory B-cell acute lymphoblastic leukemia (B-ALL) has generated great enthusiasm for the use of CAR T cells in patients with cytogenetics that portend a poor prognosis with conventional cytotoxic therapies. One such group includes infants and children with mixed lineage leukemia (MLL1, KMT2A) rearrangements (MLL-r), who fare much worse than patients with low- or standard-risk B-ALL. Although early clinical trials using CD19 CAR T cells for MLL-r B-ALL produced complete remission in most patients, relapse with CD19-negative disease was a common mechanism of treatment failure. Whereas CD19neg relapse has been observed across a broad spectrum of B-ALL patients treated with CD19-directed therapy, patients with MLL-r have manifested the emergence of AML, often clonally related to the B-ALL, suggesting that the inherent heterogeneity or lineage plasticity of MLL-r B-ALL may predispose patients to a myeloid relapse. Understanding the factors that enable and drive myeloid relapse may be important to devise strategies to improve durability of remissions. In this review, we summarize clinical observations to date with MLL-r B-ALL and generally discuss lineage plasticity as a mechanism of escape from immunotherapy.
Collapse
Affiliation(s)
- Wenjuan Liao
- Department of Pediatrics, Section of Hematology/Oncology/BMT, Center for Cancer and Blood Disorders, Children's Hospital Colorado, University of Colorado, Denver/Anschutz Medical Campus. Aurora, CO
| | - M Eric Kohler
- Department of Pediatrics, Section of Hematology/Oncology/BMT, Center for Cancer and Blood Disorders, Children's Hospital Colorado, University of Colorado, Denver/Anschutz Medical Campus. Aurora, CO
| | - Terry Fry
- Department of Pediatrics, Section of Hematology/Oncology/BMT, Center for Cancer and Blood Disorders, Children's Hospital Colorado, University of Colorado, Denver/Anschutz Medical Campus. Aurora, CO; Immunology Department and HI3 Initiative, University of Colorado, Denver/Anschutz Medical Campus. Aurora, CO
| | - Patricia Ernst
- Department of Pediatrics, Section of Hematology/Oncology/BMT, Center for Cancer and Blood Disorders, Children's Hospital Colorado, University of Colorado, Denver/Anschutz Medical Campus. Aurora, CO; Pharmacology Department, University of Colorado, Denver/Anschutz Medical Campus. Aurora, CO.
| |
Collapse
|
17
|
Jin X, Lu W, Zhang M, Xiong X, Sun R, Wei Y, He X, Zhao M. Infection Temperature Affects the Phenotype and Function of Chimeric Antigen Receptor T Cells Produced via Lentiviral Technology. Front Immunol 2021; 12:638907. [PMID: 33953713 PMCID: PMC8089475 DOI: 10.3389/fimmu.2021.638907] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/29/2021] [Indexed: 11/13/2022] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has become an important method for the treatment of hematological tumors. Lentiviruses are commonly used gene transfer vectors for preparing CAR-T cells, and the conditions for preparing CAR-T cells vary greatly. This study reported for the first time the influence of differences in infection temperature on the phenotype and function of produced CAR-T cells. Our results show that infection at 4 degrees produces the highest CAR-positive rate of T cells, infection at 37 degrees produces the fastest proliferation in CAR-T cells, and infection at 32 degrees produces CAR-T cells with the greatest proportion of naive cells and the lowest expression of immune checkpoints. Therefore, infection at 32 degrees is recommended to prepare CAR-T cells. CAR-T cells derived from infection at 32 degrees seem to have a balance between function and phenotype. Importantly, they have increased oncolytic ability. This research will help optimize the generation of CAR-T cells and improve the quality of CAR-T cell products.
Collapse
Affiliation(s)
- Xin Jin
- Nankai University School of Medicine, Tianjin, China
| | - Wenyi Lu
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Meng Zhang
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Xia Xiong
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Rui Sun
- Nankai University School of Medicine, Tianjin, China
| | - Yunxiong Wei
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Xiaoyuan He
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Mingfeng Zhao
- Nankai University School of Medicine, Tianjin, China.,Department of Hematology, Tianjin First Central Hospital, Tianjin, China.,The First Central Clinical College of Tianjin Medical University, Tianjin, China
| |
Collapse
|
18
|
Pastorczak A, Domka K, Fidyt K, Poprzeczko M, Firczuk M. Mechanisms of Immune Evasion in Acute Lymphoblastic Leukemia. Cancers (Basel) 2021; 13:1536. [PMID: 33810515 PMCID: PMC8037152 DOI: 10.3390/cancers13071536] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) results from a clonal expansion of abnormal lymphoid progenitors of B cell (BCP-ALL) or T cell (T-ALL) origin that invade bone marrow, peripheral blood, and extramedullary sites. Leukemic cells, apart from their oncogene-driven ability to proliferate and avoid differentiation, also change the phenotype and function of innate and adaptive immune cells, leading to escape from the immune surveillance. In this review, we provide an overview of the genetic heterogeneity and treatment of BCP- and T-ALL. We outline the interactions of leukemic cells in the bone marrow microenvironment, mainly with mesenchymal stem cells and immune cells. We describe the mechanisms by which ALL cells escape from immune recognition and elimination by the immune system. We focus on the alterations in ALL cells, such as overexpression of ligands for various inhibitory receptors, including anti-phagocytic receptors on macrophages, NK cell inhibitory receptors, as well as T cell immune checkpoints. In addition, we describe how developing leukemia shapes the bone marrow microenvironment and alters the function of immune cells. Finally, we emphasize that an immunosuppressive microenvironment can reduce the efficacy of chemo- and immunotherapy and provide examples of preclinical studies showing strategies for improving ALL treatment by targeting these immunosuppressive interactions.
Collapse
Affiliation(s)
- Agata Pastorczak
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, 91-738 Lodz, Poland;
| | - Krzysztof Domka
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.D.); (K.F.); (M.P.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Klaudyna Fidyt
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.D.); (K.F.); (M.P.)
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Martyna Poprzeczko
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.D.); (K.F.); (M.P.)
| | - Malgorzata Firczuk
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.D.); (K.F.); (M.P.)
| |
Collapse
|
19
|
Wiebking V, Lee CM, Mostrel N, Lahiri P, Bak R, Bao G, Roncarolo MG, Bertaina A, Porteus MH. Genome editing of donor-derived T-cells to generate allogenic chimeric antigen receptor-modified T cells: Optimizing αβ T cell-depleted haploidentical hematopoietic stem cell transplantation. Haematologica 2021; 106:847-858. [PMID: 32241852 PMCID: PMC7928014 DOI: 10.3324/haematol.2019.233882] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Indexed: 12/11/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation is an effective therapy for high-risk leukemias. In children, graft manipulation based on the selective removal of αβT cells and B cells has been shown to reduce the risk of acute and chronic graft-versus-host disease, thus allowing the use of haploidentical donors which expands the population of recipients in whom allogeneic hematopoietic stem cell transplantation can be used. Leukemic relapse, however, remains a challenge. T cells expressing chimeric antigen receptors can potently eliminate leukemia, including those in the central nervous system. We hypothesized that by engineering the donor αβT cells that are removed from the graft by genome editing to express a CD19-specific chimeric antigen receptor, while simultaneously inactivating the T-cell receptor, we could create a therapy that enhances the anti-leukemic efficacy of the stem cell transplant without increasing the risk of graft-versus-host disease. Using genome editing with Cas9 ribonucleoprotein and adeno-associated virus serotype 6, we integrated a CD19-specific chimeric antigen receptor inframe into the TRAC locus. More than 90% of cells lost T-cell receptor expression, while >75% expressed the chimeric antigen receptor. The initial product was further purified with less than 0.05% T-cell receptorpositive cells remaining. In vitro, the chimeric antigen receptor T cells efficiently eliminated target cells and produced high cytokine levels when challenged with CD19+ leukemia cells. In vivo, the gene-modified T cells eliminated leukemia without causing graft-versus-host disease in a xenograft model. Gene editing was highly specific with no evidence of off-target effects. These data support the concept that the addition of αβ T-cell-derived, genome-edited T cells expressing CD19-specific chimeric antigen receptors could enhance the anti-leukemic efficacy of αβT-celldepleted haploidentical hematopoietic stem cell transplantation without increasing the risk of graft-versus-host disease.
Collapse
Affiliation(s)
- Volker Wiebking
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Ciaran M Lee
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Nathalie Mostrel
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Premanjali Lahiri
- Laboratory for Cell and Gene Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Rasmus Bak
- Department of Biomedicine, Aarhus University, Aarhus, Denmark,Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Aarhus, Denmark
| | - Gang Bao
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Maria Grazia Roncarolo
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Alice Bertaina
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthew H Porteus
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
20
|
Simonetta F, Alam IS, Lohmeyer JK, Sahaf B, Good Z, Chen W, Xiao Z, Hirai T, Scheller L, Engels P, Vermesh O, Robinson E, Haywood T, Sathirachinda A, Baker J, Malipatlolla MB, Schultz LM, Spiegel JY, Lee JT, Miklos DB, Mackall CL, Gambhir SS, Negrin RS. Molecular Imaging of Chimeric Antigen Receptor T Cells by ICOS-ImmunoPET. Clin Cancer Res 2021; 27:1058-1068. [PMID: 33087332 PMCID: PMC7887027 DOI: 10.1158/1078-0432.ccr-20-2770] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/23/2020] [Accepted: 10/16/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Immunomonitoring of chimeric antigen receptor (CAR) T cells relies primarily on their quantification in the peripheral blood, which inadequately quantifies their biodistribution and activation status in the tissues. Noninvasive molecular imaging of CAR T cells by PET is a promising approach with the ability to provide spatial, temporal, and functional information. Reported strategies rely on the incorporation of reporter transgenes or ex vivo biolabeling, significantly limiting the application of CAR T-cell molecular imaging. In this study, we assessed the ability of antibody-based PET (immunoPET) to noninvasively visualize CAR T cells. EXPERIMENTAL DESIGN After analyzing human CAR T cells in vitro and ex vivo from patient samples to identify candidate targets for immunoPET, we employed a syngeneic, orthotopic murine tumor model of lymphoma to assess the feasibility of in vivo tracking of CAR T cells by immunoPET using the 89Zr-DFO-anti-ICOS tracer, which we have previously reported. RESULTS Analysis of human CD19-CAR T cells during activation identified the Inducible T-cell COStimulator (ICOS) as a potential target for immunoPET. In a preclinical tumor model, 89Zr-DFO-ICOS mAb PET-CT imaging detected significantly higher signal in specific bone marrow-containing skeletal sites of CAR T-cell-treated mice compared with controls. Importantly, administration of ICOS-targeting antibodies at tracer doses did not interfere with CAR T-cell persistence and function. CONCLUSIONS This study highlights the potential of ICOS-immunoPET imaging for monitoring of CAR T-cell therapy, a strategy readily applicable to both commercially available and investigational CAR T cells.See related commentary by Volpe et al., p. 911.
Collapse
Affiliation(s)
- Federico Simonetta
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University, Stanford, California
- Division of Hematology, Department of Oncology, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Translational Research Center for Oncohematology, Department of Internal Medicine Specialties, University of Geneva, Geneva, Switzerland
| | - Israt S Alam
- Bio-X Program and Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, California
| | - Juliane K Lohmeyer
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University, Stanford, California
| | - Bita Sahaf
- Stanford Cancer Institute, Stanford University, Stanford, California
| | - Zinaida Good
- Stanford Cancer Institute, Stanford University, Stanford, California
- Department of Biomedical Data Science, Stanford University, Stanford, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Weiyu Chen
- Bio-X Program and Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, California
| | - Zunyu Xiao
- Bio-X Program and Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, California
| | - Toshihito Hirai
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University, Stanford, California
| | - Lukas Scheller
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University, Stanford, California
| | - Pujan Engels
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University, Stanford, California
| | - Ophir Vermesh
- Bio-X Program and Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, California
| | - Elise Robinson
- Bio-X Program and Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, California
| | - Tom Haywood
- Bio-X Program and Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, California
| | - Ataya Sathirachinda
- Bio-X Program and Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, California
| | - Jeanette Baker
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University, Stanford, California
| | | | - Liora M Schultz
- Department of Pediatrics, Stanford University, Stanford, California
| | - Jay Y Spiegel
- Stanford Cancer Institute, Stanford University, Stanford, California
| | - Jason T Lee
- Bio-X Program and Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, California
- Stanford Cancer Institute, Stanford University, Stanford, California
- Stanford Center for Innovation in In Vivo Imaging (SCi), Stanford University School of Medicine, Stanford, California
| | - David B Miklos
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University, Stanford, California
| | - Crystal L Mackall
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University, Stanford, California
- Stanford Cancer Institute, Stanford University, Stanford, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
- Department of Pediatrics, Stanford University, Stanford, California
| | - Sanjiv S Gambhir
- Bio-X Program and Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, California
- Departments of Bioengineering and Materials Science & Engineering, Bio-X, Stanford University, Stanford, California
| | - Robert S Negrin
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University, Stanford, California.
| |
Collapse
|
21
|
Ishii K, Pouzolles M, Chien CD, Erwin-Cohen RA, Kohler ME, Qin H, Lei H, Kuhn S, Ombrello AK, Dulau-Florea A, Eckhaus MA, Shalabi H, Yates B, Lichtenstein DA, Zimmermann VS, Kondo T, Shern JF, Young HA, Taylor N, Shah NN, Fry TJ. Perforin-deficient CAR T cells recapitulate late-onset inflammatory toxicities observed in patients. J Clin Invest 2020; 130:5425-5443. [PMID: 32925169 PMCID: PMC7524496 DOI: 10.1172/jci130059] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/09/2020] [Indexed: 12/20/2022] Open
Abstract
Late-onset inflammatory toxicities resembling hemophagocytic lymphohistiocytosis (HLH) or macrophage activation syndrome (MAS) occur after chimeric antigen receptor T cell (CAR T cell) infusion and represent a therapeutic challenge. Given the established link between perforin deficiency and primary HLH, we investigated the role of perforin in anti-CD19 CAR T cell efficacy and HLH-like toxicities in a syngeneic murine model. Perforin contributed to both CD8+ and CD4+ CAR T cell cytotoxicity but was not required for in vitro or in vivo leukemia clearance. Upon CAR-mediated in vitro activation, perforin-deficient CAR T cells produced higher amounts of proinflammatory cytokines compared with WT CAR T cells. Following in vivo clearance of leukemia, perforin-deficient CAR T cells reexpanded, resulting in splenomegaly with disruption of normal splenic architecture and the presence of hemophagocytes, which are findings reminiscent of HLH. Notably, a substantial fraction of patients who received anti-CD22 CAR T cells also experienced biphasic inflammation, with the second phase occurring after the resolution of cytokine release syndrome, resembling clinical manifestations of HLH. Elevated inflammatory cytokines such as IL-1β and IL-18 and concurrent late CAR T cell expansion characterized the HLH-like syndromes occurring in the murine model and in humans. Thus, a murine model of perforin-deficient CAR T cells recapitulated late-onset inflammatory toxicities occurring in human CAR T cell recipients, providing therapeutically relevant mechanistic insights.
Collapse
Affiliation(s)
- Kazusa Ishii
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, and
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, USA
| | - Marie Pouzolles
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH
| | - Christopher D. Chien
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH
| | - Rebecca A. Erwin-Cohen
- Cancer and Inflammation Program, Center for Cancer Research, NCI, NIH, Frederick, Maryland, USA
| | - M. Eric Kohler
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH
- Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Haiying Qin
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH
| | - Haiyan Lei
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH
| | - Skyler Kuhn
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, NCI, NIH, Bethesda, Maryland, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Amanda K. Ombrello
- Inflammatory Disease Section, National Human Genome Research Institute, NIH
| | | | - Michael A. Eckhaus
- Diagnostic and Research Services Branch, Division of Veterinary Resources, NIH, Bethesda, Maryland, USA
| | - Haneen Shalabi
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH
| | - Bonnie Yates
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH
| | - Daniel A. Lichtenstein
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH
| | - Valérie S. Zimmermann
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH
- Université de Montpellier, IGMM, CNRS, Montpellier, France
| | - Taisuke Kondo
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH
| | - Jack F. Shern
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH
| | - Howard A. Young
- Cancer and Inflammation Program, Center for Cancer Research, NCI, NIH, Frederick, Maryland, USA
- Laboratory of Cancer Immunometabolism, Center for Cancer Research, NCI, NIH, Frederick, Maryland, USA
| | - Naomi Taylor
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH
- Université de Montpellier, IGMM, CNRS, Montpellier, France
| | - Nirali N. Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH
| | - Terry J. Fry
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH
- Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado, USA
| |
Collapse
|
22
|
Hutt D, Bielorai B, Baturov B, Z'orbinski I, Ilin N, Adam E, Itzhaki O, Besser MJ, Toren A, Jacoby E. Feasibility of leukapheresis for CAR T-cell production in heavily pre-treated pediatric patients. Transfus Apher Sci 2020; 59:102769. [PMID: 32414613 DOI: 10.1016/j.transci.2020.102769] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/26/2020] [Accepted: 03/26/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Autologous CD19 chimeric-antigen receptor (CAR) T-cells are an effective salvage therapy for patients with relapsed or refractory B cell malignancies. The essential first step in the production is the collection of mature lymphocytes through leukapheresis. It is a challenging procedure given the fact patients are heavily pretreated and the special considerations of pediatric apheresis. METHODS We analyzed the data of leukapheresis outcome for CAR T production in a phase 1b/2 clinical trial enrolling 34 children, adolescents and young adults with relapsed or refractory B-cell malignancies. RESULTS All patients underwent a single leukapheresis. Given a short production time for CAR T-cells, most patients received bridging therapy prior to apheresis. Leukapheresis was performed using peripheral venous access in the majority (82%) of patients, and the remainder required arterial line or central venous access. T-cell collection efficiency (CE) was variable with a median of 18%. No apheresis-related adverse events were noted, and all procedures were successful but two: one resulting in lower than target dose (1 × 106 CAR + cells/kg) and the other in failure of CAR T-cell production. CONCLUSIONS Collection of sufficient T-cells in heavily pretreated pediatric patients via a single apheresis procedure is feasible even with relatively low T-cell CE.
Collapse
Affiliation(s)
- Daphna Hutt
- Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Bella Bielorai
- Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Bella Baturov
- Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Inna Z'orbinski
- Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Natalia Ilin
- Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Etai Adam
- Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Orit Itzhaki
- Ella Institute of Immuno-Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Michal J Besser
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Ella Institute of Immuno-Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Amos Toren
- Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Elad Jacoby
- Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
23
|
Blaeschke F, Willier S, Stenger D, Lepenies M, Horstmann MA, Escherich G, Zimmermann M, Rojas Ringeling F, Canzar S, Kaeuferle T, Rohlfs M, Binder V, Klein C, Feuchtinger T. Leukemia-induced dysfunctional TIM-3 +CD4 + bone marrow T cells increase risk of relapse in pediatric B-precursor ALL patients. Leukemia 2020; 34:2607-2620. [PMID: 32203137 DOI: 10.1038/s41375-020-0793-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 02/29/2020] [Accepted: 03/03/2020] [Indexed: 12/17/2022]
Abstract
Interaction of malignancies with tissue-specific immune cells has gained interest for prognosis and intervention of emerging immunotherapies. We analyzed bone marrow T cells (bmT) as tumor-infiltrating lymphocytes in pediatric precursor-B cell acute lymphoblastic leukemia (ALL). Based on data from 100 patients, we show that ALL is associated with late-stage CD4+ phenotype and loss of early CD8+ T cells. The inhibitory exhaustion marker TIM-3 on CD4+ bmT increased relapse risk (RFS = 94.6/70.3%) confirmed by multivariate analysis. The hazard ratio of TIM-3 expression nearly reached the hazard ratio of MRD (7.1 vs. 8.0) indicating that patients with a high frequency of TIM-3+CD4+ bone marrow T cells at initial diagnosis have a 7.1-fold increased risk to develop ALL relapse. Comparison of wild type primary T cells to CRISPR/Cas9-mediated TIM-3 knockout and TIM-3 overexpression confirmed the negative effect of TIM-3 on T cell responses against ALL. TIM-3+CD4+ bmT are increased in ALL overexpressing CD200, that leads to dysfunctional antileukemic T cell responses. In conclusion, TIM-3-mediated interaction between bmT and leukemia cells is shown as a strong risk factor for relapse in pediatric B-lineage ALL. CD200/TIM-3-signaling, rather than PD-1/PD-L1, is uncovered as a mechanism of T cell dysfunction in ALL with major implication for future immunotherapies.
Collapse
Affiliation(s)
- Franziska Blaeschke
- Dr. von Hauner Children's Hospital, University Hospital, Ludwig Maximilian University, 80337, Munich, Germany
| | - Semjon Willier
- Dr. von Hauner Children's Hospital, University Hospital, Ludwig Maximilian University, 80337, Munich, Germany
| | - Dana Stenger
- Dr. von Hauner Children's Hospital, University Hospital, Ludwig Maximilian University, 80337, Munich, Germany
| | - Mareike Lepenies
- Dr. von Hauner Children's Hospital, University Hospital, Ludwig Maximilian University, 80337, Munich, Germany
| | - Martin A Horstmann
- Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Gabriele Escherich
- Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Martin Zimmermann
- Department of Pediatric Hematology and Oncology, Hannover Medical School, 30625, Hannover, Germany
| | | | - Stefan Canzar
- Gene Center, Ludwig Maximilian University Munich, 81377, Munich, Germany
| | - Theresa Kaeuferle
- Dr. von Hauner Children's Hospital, University Hospital, Ludwig Maximilian University, 80337, Munich, Germany
| | - Meino Rohlfs
- Dr. von Hauner Children's Hospital, University Hospital, Ludwig Maximilian University, 80337, Munich, Germany
| | - Vera Binder
- Dr. von Hauner Children's Hospital, University Hospital, Ludwig Maximilian University, 80337, Munich, Germany
| | - Christoph Klein
- Dr. von Hauner Children's Hospital, University Hospital, Ludwig Maximilian University, 80337, Munich, Germany.,Gene Center, Ludwig Maximilian University Munich, 81377, Munich, Germany
| | - Tobias Feuchtinger
- Dr. von Hauner Children's Hospital, University Hospital, Ludwig Maximilian University, 80337, Munich, Germany.
| |
Collapse
|
24
|
Differential Depletion of Bone Marrow Resident B-ALL after Systemic Administration of Endosomal TLR Agonists. Cancers (Basel) 2020; 12:cancers12010169. [PMID: 32015298 PMCID: PMC7016792 DOI: 10.3390/cancers12010169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/04/2020] [Accepted: 01/08/2020] [Indexed: 12/14/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common pediatric malignancy. While frontline chemotherapy regimens are generally very effective, the prognosis for patients whose leukemia returns remains poor. The presence of measurable residual disease (MRD) in bone marrow at the completion of induction therapy is the strongest predictor of relapse, suggesting that strategies to eliminate the residual leukemic blasts from this niche could reduce the incidence of recurrence. We have previously reported that toll-like receptor (TLR) agonists achieve durable T cell-mediated protection in transplantable cell line-based models of B cell precursor leukemia (B-ALL). However, the successful application of TLR agonist therapy in an MRD setting would require the induction of anti-leukemic immune activity specifically in the bone marrow, a site of the chemotherapy-resistant leukemic blasts. In this study, we compare the organ-specific depletion of human and mouse primary B-ALL cells after systemic administration of endosomal TLR agonists. Despite comparable splenic responses, only the TLR9 agonist induced strong innate immune responses in the bone marrow and achieved a near-complete elimination of B-ALL cells. This pattern of response was associated with the most significantly prolonged disease-free survival. Overall, our findings identify innate immune activity in the bone marrow that is associated with durable TLR-induced protection against B-ALL outgrowth.
Collapse
|
25
|
Maffini E, Saraceni F, Lanza F. Treatment of Adult Patients with Relapsed/Refractory B-Cell Philadelphia-Negative Acute Lymphoblastic Leukemia. Clin Hematol Int 2019; 1:85-93. [PMID: 34595415 PMCID: PMC8432388 DOI: 10.2991/chi.d.190503.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 04/26/2019] [Indexed: 12/27/2022] Open
Abstract
The majority of adult patients affected by B-cell acute lymphoblastic leukemia (B-ALL) will relapse after an initial response, while approximately 20% will display primary resistant disease. Patients suffering from relapsed/refractory B-ALL have a very poor outcome. Allogeneic hematopoietic cell transplantation (HCT) still represents the only curative approach, but is not so frequently feasible, because of patient's fitness, donor availability, and the ability to achieve a remission prior to HCT. The estimated remission rates with conventional cytotoxic agents are around 30%, but they are short-lived. These disappointing results led to the introduction of new immunologic-based treatments-blinatumomab and inotuzumab. They produced a substantial improvement in terms of response rates, with the ability, in most cases, to induce a minimal residual disease (MRD)-negative status. Similarly, T cells engineered to express a CD19-specific chimeric antigen receptor (CAR-T) have yielded sensational results among patients with relapsed/refractory B-ALL, with unexpectedly high MRD-negative complete remissions rates. However, the first studies looking at long-term outcomes after CAR-T infusions told us that a significant fraction of such responses are not durable, and may benefit from a consolidation approach such as an allogeneic HCT.
Collapse
Affiliation(s)
- Enrico Maffini
- Hematology Unit, Romagna Transplant Network, Hospital of Ravenna, Viale Randi n. 5, 48121 Ravenna, Italy
| | - Francesco Saraceni
- Hematology Unit, Romagna Transplant Network, Hospital of Ravenna, Viale Randi n. 5, 48121 Ravenna, Italy
| | - Francesco Lanza
- Hematology Unit, Romagna Transplant Network, Hospital of Ravenna, Viale Randi n. 5, 48121 Ravenna, Italy
| |
Collapse
|
26
|
Li T, Zhang Y, Peng D, Mao X, Zhou X, Zhou J. A good response of refractory mantel cell lymphoma to haploidentical CAR T cell therapy after failure of autologous CAR T cell therapy. J Immunother Cancer 2019; 7:51. [PMID: 30791947 PMCID: PMC6383265 DOI: 10.1186/s40425-019-0529-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 02/06/2019] [Indexed: 12/31/2022] Open
Abstract
Background The aggressive form of Mantle cell non-hodgkin B cell lymphoma (MCL) has a dismal prognosis. Dual targeting BTK and BCL2 with ibrutinib and venetoclax has improved outcomes in MCL patients who were predicted not to respond to conventional therapy, but it is unlikely to be curative. Chimeric antigen receptor-modified T (CAR T) cells exhibit very effective function in elimination of relapsed/refractory B-cell lymphoid malignancies, we investigated their use in a patient with relapsed MCL. Case presentation Here, we report a case of a refractory MCL in a patient who had relapsed after conventional chemotherapy and autologous CAR T cell therapy. The patient received multiple molecularly targeted therapies, including targeting BTK and BCL2, and haplo-identical CAR T (haplo-CAR T) cells from her daughter without previous allo-hematopoietic stem cell transplantation. Haplo-CAR T cells could effectively proliferate in vivo and had a clinically significant antitumor activity without serious side effects. The patient achieved a partial remission, with minimal residual disease. Conclusions This case suggests that haplo-CAR T cell therapy can be effective in controlling lymphoma that failed to respond to autologous CAR T cell therapy and overcome limitation of autologous CAR T cells, thus may be one possible regimen before the era of off-the-shelf “universal” CAR T cell therapy. Trial registration ChiCTR-OPN-16008526. http://www.chictr.org.cn/showproj.aspx?proj=13798; ChiCTR1800019385. http://www.chictr.org.cn/showproj.aspx?proj=32805; ChiCTR1800019449. http://www.chictr.org.cn/showproj.aspx?proj=32778. Electronic supplementary material The online version of this article (10.1186/s40425-019-0529-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tongjuan Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuanyuan Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dan Peng
- Department of Nuclear medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xia Mao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoxi Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|