1
|
Patel KK, Tariveranmoshabad M, Kadu S, Shobaki N, June C. From concept to cure: The evolution of CAR-T cell therapy. Mol Ther 2025; 33:2123-2140. [PMID: 40070120 DOI: 10.1016/j.ymthe.2025.03.005] [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: 01/28/2025] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/21/2025] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has revolutionized cancer immunotherapy in the 21st century, providing innovative solutions and life-saving therapies for previously untreatable diseases. This approach has shown remarkable success in treating various hematological malignancies and is now expanding into clinical trials for solid tumors, such as prostate cancer and glioblastoma, as well as infectious and autoimmune diseases. CAR-T cell therapy involves harvesting a patient's T cells, genetically engineering them with viral vectors to express CARs targeting specific antigens and reinfusing the modified cells into the patient. These CAR-T cells function independently of major histocompatibility complex (MHC) antigen presentation, selectively identifying and eliminating target cells. This review highlights the key milestones in CAR-T cell evolution, from its invention to its clinical applications. It outlines the historical timeline leading to the invention of CAR-T cells, discusses the major achievements that have transformed them into a breakthrough therapy, and addresses remaining challenges, including high manufacturing costs, limited accessibility, and toxicity issues such as cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. Additionally, the review explores future directions and advances in the field, such as developing next-generation CAR-T cells aiming to maximize efficacy, minimize toxicity, and broaden therapeutic applications.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/trends
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Neoplasms/therapy
- Neoplasms/immunology
- Animals
Collapse
Affiliation(s)
- Kisha K Patel
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mito Tariveranmoshabad
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Siddhant Kadu
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nour Shobaki
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl June
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Cadinanos-Garai A, Flugel CL, Cheung A, Jiang E, Vaissié A, Abou-El-Enein M. High-dimensional temporal mapping of CAR T cells reveals phenotypic and functional remodeling during manufacturing. Mol Ther 2025; 33:2291-2309. [PMID: 40315840 DOI: 10.1016/j.ymthe.2025.04.006] [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: 03/04/2025] [Revised: 04/02/2025] [Accepted: 04/02/2025] [Indexed: 05/04/2025] Open
Abstract
Despite the notable success of chimeric antigen receptor (CAR) T cell therapies in hematological malignancies, clinical outcomes remain variable, making it critical to understand how manufacturing influences product composition and function. We developed a 36-marker spectral flow cytometry panel enabling integrated profiling of phenotypic, metabolic, and functional attributes across CAR T cell production. Mid-expansion products (day 5) retained stem-like, metabolically active CD4+ Th1 subsets with high proliferative capacity, whereas prolonged culture (day 10) enriched terminally differentiated CD8+ Tc1 cells and NK-like T cell populations. CAR+ and CAR- T cells showed similar differentiation trajectories, suggesting that culture conditions may have a larger impact on phenotypic remodeling than CAR integration. Upon antigen encounter and restimulation, day 5 and day 10 products showed comparable cytotoxicity, while differing in their activation and checkpoint profiles. Cryopreservation modestly affected stem cell memory, activation, and metabolic markers but preserved overall phenotype and cytotoxic function. These findings establish a high-dimensional framework for mapping CAR T cell dynamics to support manufacturing optimization and next-generation cell therapy design.
Collapse
Affiliation(s)
- Amaia Cadinanos-Garai
- USC/CHLA Cell Therapy Program, University of Southern California and Children's Hospital of Los Angeles, Los Angeles, CA 90033, USA
| | - Christian L Flugel
- USC/CHLA Cell Therapy Program, University of Southern California and Children's Hospital of Los Angeles, Los Angeles, CA 90033, USA; Berlin Center for Advanced Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
| | - Anson Cheung
- USC/CHLA Cell Therapy Program, University of Southern California and Children's Hospital of Los Angeles, Los Angeles, CA 90033, USA
| | - Enzi Jiang
- USC/CHLA Cell Therapy Program, University of Southern California and Children's Hospital of Los Angeles, Los Angeles, CA 90033, USA
| | - Alix Vaissié
- USC/CHLA Cell Therapy Program, University of Southern California and Children's Hospital of Los Angeles, Los Angeles, CA 90033, USA
| | - Mohamed Abou-El-Enein
- USC/CHLA Cell Therapy Program, University of Southern California and Children's Hospital of Los Angeles, Los Angeles, CA 90033, USA; Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Regulatory and Quality Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
3
|
DiAndreth B, Nesterenko PA, Winters AG, Flynn AD, Jette CA, Suryawanshi V, Shafaattalab S, Martire S, Daris M, Moore E, Elshimali R, Gill T, Riley TP, Miller S, Netirojjanakul C, Hamburger AE, Kamb A. Multi-targeted, NOT gated CAR-T cells as a strategy to protect normal lineages for blood cancer therapy. Front Immunol 2025; 16:1493329. [PMID: 40191207 PMCID: PMC11968376 DOI: 10.3389/fimmu.2025.1493329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 02/21/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Despite advances in treatment of blood cancers, several-including acute myeloid leukemia (AML)-continue to be recalcitrant. Cell therapies based on chimeric antigen receptors (CARs) have emerged as promising approaches for blood cancers. However, current CAR-T treatments suffer from on-target, off-tumor toxicity, because most familiar blood cancer targets are also expressed in normal lineages. In addition, they face the common problem of relapse due to target-antigen loss. Cell therapeutics engineered to integrate more than one signal, often called logic-gated cells, can in principle achieve greater selectivity for tumors. Methods We applied such a technology, a NOT gated system called Tmod™ that is being developed to treat solid-tumor patients, to the problem of therapeutic selectivity for blood cancer cells. Results Here we show that Tmod cells can be designed to target 2-4 antigens to provide different practical and conceptual options for a blood cancer therapy: (i) mono- and bispecific activating receptors that target CD33, a well-known AML antigen expressed on the majority of AML tumors (as well as healthy myeloid cells) and CD43 (SPN), an antigen expressed on many hematopoietic cancers (and normal blood lineages); and (ii) mono- and bispecific inhibitory receptors that target CD16b (FCGR3B) and CLEC9A, antigens expressed on key normal blood cells but not on most blood cancers. Discussion These results further demonstrate the robust modularity of the Tmod system and generalize the Tmod approach beyond solid tumors.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Animals
- Hematologic Neoplasms/therapy
- Hematologic Neoplasms/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- T-Lymphocytes/metabolism
- Mice
- Antigens, Neoplasm/immunology
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Cell Line, Tumor
- Cell Lineage
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Alexander Kamb
- A2 Biotherapeutics Discovery Research, Agoura Hills, CA, United States
| |
Collapse
|
4
|
Newsam AD, Ziccheddu B, Gowda Saralamma VV, Coughlin CA, Goretsky YE, Youssfi AA, Russo MV, Gallego NC, Fattakhov N, Coffey DG, Tsai DE, Carmona-Berrio D, Suissa DM, Manara P, Sondhi AK, Roberts ER, Sheffield-Veney I, Spiegel JY, Amador C, Alderuccio JP, Bilbao D, Jain MD, Maura F, Locke FL, Schatz JH. RHOA Loss of Function Impairs the IFNγ Response and Promotes CD19 Antigen Escape to Drive CAR-T Resistance in Diffuse Large B-cell Lymphoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640687. [PMID: 40093149 PMCID: PMC11908125 DOI: 10.1101/2025.02.27.640687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
CD19-directed chimeric antigen receptor (CAR)-T cells are breakthrough therapies for aggressive B-cell lymphomas, but less than half of patients achieve durable responses. We previously showed through whole-genome sequencing of tumors from CAR-T-treated patients that deletions of RHOA (3p21.31) are enriched in cases progressing after treatment. RHOA 's roles in resistance and pathogenesis are poorly defined, despite loss-of-function alterations that occur in ~20% of newly diagnosed diffuse large B-cell lymphoma (DLBCL) cases. To evaluate mechanisms of CAR-T resistance, we created RHOA-deficient DLBCL systems and confirmed cell-intrinsic loss of response to CAR-19 in vitro and in vivo. RHOA loss promotes AKT activation that impairs cell-intrinsic responses to interferon gamma (IFNγ). Moreover, expression of the CAR target CD19 is consistently down-regulated accompanied by a drive toward plasmablast differentiation. RHOA deficient tumors demonstrate greatly increased sensitivity to AKT-pathway inhibitors, which reverse impaired IFNγ responses. Lymphoma microenvironments in vivo in immunocompetent mice reveal that RHOA loss promotes decreased infiltration by cytotoxic T cells and enrichment of M2-polarized macrophages, known markers of CAR-T resistance in lymphoma clinical cases. Overall, we characterize RHOA deficiency as an AKT-mediated CAR-T resistance driver and implicate avoidance of T-cell mediated killing as a likely reason for RHOA's frequent loss in DLBCL pathogenesis.
Collapse
|
5
|
Ye X, Ge M, Tan M, Wu Y, Zhang H, Fu Z. CD19 -targeted CAR T therapy treating hematologic malignancies: hidden danger is the next neighbor to security? Front Immunol 2025; 16:1490491. [PMID: 40103829 PMCID: PMC11914092 DOI: 10.3389/fimmu.2025.1490491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 02/18/2025] [Indexed: 03/20/2025] Open
Abstract
CD19-targeted chimeric antigen receptor (CAR) T-cell therapy has achieved marvelous results in the treatment of patients with relapsed and/or refractory B-cell lymphomas, B-cell acute lymphoblastic leukemia, and multiple myeloma. As a new treatment method that has changed the existing treatment paradigm, there has been a short time from its emergence to FDA approval. However, with the increasing number of cases and the passage of time, hidden problems have gradually been exposed. In this review, we summarize the short- and long-term toxicity, such as secondary T-cell tumors and lethal CAR tumors, of patients with hematologic malignancies treated with CD19-CAR-T cells, including cytokine release syndrome (CRS), ICANS, and secondary malignancies with low occurrence rates but high mortality, such as secondary T cell tumors and lethal CAR tumors, which may be related to the gene modification mechanism of viral vectors currently approved for CAR-T cells. We also discuss potential investigational strategies designed to improve the safety of CAR-T-cell therapy.
Collapse
Affiliation(s)
- Xueshuai Ye
- Affiliated Hospital of Hebei Engineering University and School of Clinical Medicine, Hebei University of Engineering, Handan, China
| | - Min Ge
- Affiliated Hospital of Hebei Engineering University and School of Clinical Medicine, Hebei University of Engineering, Handan, China
| | - Mengtian Tan
- Affiliated Hospital of Hebei Engineering University and School of Clinical Medicine, Hebei University of Engineering, Handan, China
| | - Yongqiang Wu
- Gene Editing Research Center, Hebei University of Science and Technology, Shijiazhuang, China
| | - Haiqiang Zhang
- Department of Gastrointestinal Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zexian Fu
- Medical College, Hebei University of Engineering, Handan, China
| |
Collapse
|
6
|
de Oliveira Canedo G, Roddie C, Amrolia PJ. Dual-targeting CAR T cells for B-cell acute lymphoblastic leukemia and B-cell non-Hodgkin lymphoma. Blood Adv 2025; 9:704-721. [PMID: 39631066 PMCID: PMC11869864 DOI: 10.1182/bloodadvances.2024013586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/24/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024] Open
Abstract
ABSTRACT Relapse after CD19-directed chimeric antigen receptor (CAR) T-cell therapy remains a major challenge in B-cell acute lymphoblastic leukemia (ALL) and B-cell non-Hodgkin lymphoma (B-NHL). One of the main strategies to avoid CD19-negative relapse has been the development of dual CAR T cells targeting CD19 and an additional target, such as CD22 or CD20. Different methods have been used to achieve this, including coadministration of 2 products targeting 1 single antigen, cotransduction of autologous T cells, use of a bicistronic vector, or the development of bivalent CARs. Phase 1 and 2 trials across all manufacturing strategies have shown this to be a safe approach with equivalent remission rates and initial product expansion. CAR T-cell persistence remains a significant issue, with the majority of relapses being antigen-positive after CAR T-cell infusion. Further, despite adding a second antigen, antigen-negative relapses have not yet been eliminated. This review summarizes the state of the art with dual-targeting CAR T cells for B-cell ALL and B-NHL, the challenges encountered, and possible next steps to overcome them.
Collapse
Affiliation(s)
- Gustavo de Oliveira Canedo
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
- Department of Bone Marrow Transplant, Great Ormond Street Hospital, London, United Kingdom
| | - Claire Roddie
- Department of Haematology, University College London Hospitals, London, United Kingdom
| | - Persis J. Amrolia
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
- Department of Bone Marrow Transplant, Great Ormond Street Hospital, London, United Kingdom
| |
Collapse
|
7
|
Zhang B, Wu J, Jiang H, Zhou M. Strategies to Overcome Antigen Heterogeneity in CAR-T Cell Therapy. Cells 2025; 14:320. [PMID: 40072049 PMCID: PMC11899321 DOI: 10.3390/cells14050320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/09/2025] [Accepted: 02/18/2025] [Indexed: 03/15/2025] Open
Abstract
Chimeric antigen receptor (CAR) gene-modified T-cell therapy has achieved significant success in the treatment of hematological malignancies. However, this therapy has not yet made breakthroughs in the treatment of solid tumors and still faces issues of resistance and relapse in hematological cancers. A major reason for these problems is the antigenic heterogeneity of tumor tissues. This review outlines the antigenic heterogeneity encountered in CAR-T cell therapy and the corresponding strategies to address it. These strategies include using combination therapy to increase the abundance of target antigens, optimizing the structure of CARs to enhance sensitivity to low-density antigens, developing multi-targeted CAR-T cells, and reprogramming the TME to activate endogenous immunity. These approaches offer new directions for overcoming tumor antigenic heterogeneity in CAR-T cell therapy.
Collapse
Affiliation(s)
- Bohan Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China; (B.Z.); (J.W.)
| | - Jiawen Wu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China; (B.Z.); (J.W.)
| | - Hua Jiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China; (B.Z.); (J.W.)
- CARsgen Therapeutics, Shanghai 200231, China
| | - Min Zhou
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China; (B.Z.); (J.W.)
| |
Collapse
|
8
|
Zhang C, Liu H. Advancements and Future Directions of Dual-Target Chimeric Antigen Receptor T-Cell Therapy in Preclinical and Clinical Studies. J Immunol Res 2025; 2025:5845167. [PMID: 39844819 PMCID: PMC11753851 DOI: 10.1155/jimr/5845167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 12/18/2024] [Indexed: 01/24/2025] Open
Abstract
In recent years, chimeric antigen receptor T-cell (CAR-T) therapy has made groundbreaking progress in the treatment of various cancer types, particularly hematological malignancies. In the meantime, various preclinical and clinical studies have extensively explored dual-target CAR-T therapies which can be designed to recognize two antigens simultaneously based on the immunophenotype of tumor cells. Compared with single-target CAR-T approach, dual-target CAR-T therapies demonstrate varying degrees of superior antitumor CAR effects, prevent antigen escape and relapse, reduce on-target off-tumor effects, and ensure durable responses in different types of cancer. These advantages highlight the potential future prospects in this field, showing varying degrees of advancement in preclinical and clinical studies. Herein, we aimed to review different dual-target CAR-T studies conducted on a wide range of tumor models, summarizing the selection of target combinations, the efficacy and safety demonstrated in preclinical and clinical settings, the existing limitations, and the potential future directions of this promising therapeutic strategy.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/trends
- Immunotherapy, Adoptive/adverse effects
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Animals
- Neoplasms/therapy
- Neoplasms/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/transplantation
- Antigens, Neoplasm/immunology
- Clinical Trials as Topic
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
- Treatment Outcome
Collapse
Affiliation(s)
- Chenyun Zhang
- School of Medicine, University of Tsinghua, Beijing, China
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Haizhou Liu
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
9
|
Agliardi G, Dias J, Rampotas A, Garcia J, Roddie C. Accelerating and optimising CAR T-cell manufacture to deliver better patient products. Lancet Haematol 2025; 12:e57-e67. [PMID: 39510106 DOI: 10.1016/s2352-3026(24)00273-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/17/2024] [Accepted: 08/20/2024] [Indexed: 11/15/2024]
Abstract
Autologous chimeric antigen receptor (CAR) T-cell therapy has transformed the management of B-cell leukaemia and lymphoma. However, current manufacturing processes present logistical hurdles, restricting broader application. As clinical outcomes can be heavily influenced by the quality of autologous starting materials and production processes, strategies to improve product phenotype are crucial. Short manufacturing processes have the advantage of bringing products to patients more quickly and, in parallel, avoiding the highly differentiated and exhausted CAR T-cell phenotypes associated with prolonged ex vivo manufacture. This Review examines advances in our understanding of what constitutes an effective CAR T-cell product and approaches to improve product quality. Historically, strategies have relied on adjustments in medium composition and selection of less differentiated cell subtypes. Since 2020, the field has been shifting towards reduced-expansion protocols, no-activation protocols, and point-of-care manufacturing. These approaches have the advantage of a rapid turnaround while maintaining a less differentiated and exhausted phenotype. These efforts are leading to ultrarapid production methods and even elimination of ex vivo manipulation with the use of in vivo manufacturing approaches. In this Review, we focus on the advances needed to accelerate CAR T-cell manufacture (including near-patient methods), with an emphasis on improved therapeutic efficacy and rapid turnaround time, and simplified quality control procedures required to fully realise the clinical potential of CAR T-cell therapies.
Collapse
Affiliation(s)
- Giulia Agliardi
- Cancer Institute, University College London, London, UK; Centre for Cell, Gene and Tissue Therapeutics, Royal Free Hospital London, NHS Foundation Trust, London, UK
| | - Juliana Dias
- Cancer Institute, University College London, London, UK; Centre for Cell, Gene and Tissue Therapeutics, Royal Free Hospital London, NHS Foundation Trust, London, UK
| | - Alexandros Rampotas
- Cancer Institute, University College London, London, UK; Department of Haematology, University College London Hospitals, London, UK
| | - John Garcia
- Cancer Institute, University College London, London, UK; Centre for Cell, Gene and Tissue Therapeutics, Royal Free Hospital London, NHS Foundation Trust, London, UK
| | - Claire Roddie
- Cancer Institute, University College London, London, UK; Department of Haematology, University College London Hospitals, London, UK.
| |
Collapse
|
10
|
Esquinas E, Moreno-Sanz A, Sandá V, Stodulski-Ciesla D, Borregón J, Peña-Blanque V, Fernández-Calles J, Fernandez-Fuentes N, Serrano-Lopez J, Juan M, Engel P, Llamas-Sillero P, Solán-Blanco L, Martin-Antonio B. Preclinical development of three novel CARs targeting CD79b for the treatment of non-Hodgkin's lymphoma and characterization of the loss of the target antigen. J Immunother Cancer 2024; 12:e009485. [PMID: 39694704 PMCID: PMC11667269 DOI: 10.1136/jitc-2024-009485] [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: 04/17/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Infusion of T cells modified with a chimeric antigen receptor (CAR) targeting CD19 has achieved exceptional responses in patients with non-Hodgkin's lymphoma (NHL), which led to the approval of CAR targeting CD19 (CART19) (Axi-cel and Liso-cel) as second line of treatment for adult patients with relapsed/refractory NHL. Unfortunately, 60% of patients still relapse after CART19 due to either a loss of expression of the target antigen (CD19) in the tumor cell, observed in 27% of relapsed patients, a limited CAR-T persistence, and additional mechanisms, including the suppression of the tumor microenvironment. Clinic strategies to prevent target antigen loss include sequential treatment with CARs directed at CD20 or CD22, which have caused loss of the second antigen, suggesting targeting other antigens less prone to disappear. CD79b, expressed in NHL, is a target in patients treated with antibody-drug conjugates (ADC). However, the limited efficacy of ADC suggests that a CAR therapy targeting CD79b might improve results. METHODS We designed three new CARs against CD79b termed CAR for Lymphoma (CARLY)1, 2 and 3. We compared their efficacy, phenotype, and inflammatory profiles with CART19 (ARI0001) and CARTBCMA (ARI0002h), which can treat NHL. We also analyzed the target antigen's expression loss (CD79b, CD19, and B-cell maturation antigen(BCMA)). RESULTS We found that CARLY2 and CARLY3 had high affinity and specificity towards CD79b on B cells. In vitro, all CAR-T cells had similar anti-NHL efficacy, which was retained in an NHL model of CD19- relapse. In vivo, CARLY3 showed the highest efficacy. Analysis of the loss of the target antigen demonstrated that CARLY cells induced CD79b and CD19 downregulation on NHL cells with concomitant trogocytosis of these antigens to T cells, being most notorious in CARLY2, which had the highest affinity towards CD79b and CD19, and supporting the selection of CARLY3 to design a new treatment for patients with NHL. Finally, we created a CAR treatment based on dual targeting of CD79b and BCMA to avoid losing the target antigen. This treatment showed the highest efficacy and did not cause loss of the target antigen. CONCLUSIONS Based on specificity, efficacy, and loss of the target antigen, CARLY3 represents a potential novel CAR treatment for NHL.
Collapse
Affiliation(s)
- Esperanza Esquinas
- Department of Experimental Hematology, Health Research Institute of the Jimenez Diaz Foundation, UAM, Madrid, Spain, UAM, Madrid, Spain
- Next Generation CART MAD Consortium, Madrid, Spain
- Departamento de Desarrollo de Medicamentos de Terapias Avanzadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Alvaro Moreno-Sanz
- Department of Experimental Hematology, Health Research Institute of the Jimenez Diaz Foundation, UAM, Madrid, Spain, UAM, Madrid, Spain
| | - Victor Sandá
- Department of Experimental Hematology, Health Research Institute of the Jimenez Diaz Foundation, UAM, Madrid, Spain, UAM, Madrid, Spain
| | - Damian Stodulski-Ciesla
- Department of Experimental Hematology, Health Research Institute of the Jimenez Diaz Foundation, UAM, Madrid, Spain, UAM, Madrid, Spain
| | - Jennifer Borregón
- Department of Experimental Hematology, Health Research Institute of the Jimenez Diaz Foundation, UAM, Madrid, Spain, UAM, Madrid, Spain
| | - Virginia Peña-Blanque
- Department of Immunology, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, UAM, Madrid, Spain
| | - Javier Fernández-Calles
- Department of Biomedical Science, University of Barcelona Faculty of Medicine and Health Sciences, Barcelona, Spain
| | | | - Juana Serrano-Lopez
- Department of Experimental Hematology, Health Research Institute of the Jimenez Diaz Foundation, UAM, Madrid, Spain, UAM, Madrid, Spain
- Next Generation CART MAD Consortium, Madrid, Spain
| | - Manel Juan
- Hospital Clínic de Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, Spain
| | - Pablo Engel
- Department of Biomedical Science, University of Barcelona Faculty of Medicine and Health Sciences, Barcelona, Spain
| | - Pilar Llamas-Sillero
- Department of Experimental Hematology, Health Research Institute of the Jimenez Diaz Foundation, UAM, Madrid, Spain, UAM, Madrid, Spain
- Next Generation CART MAD Consortium, Madrid, Spain
| | - Laura Solán-Blanco
- Department of Experimental Hematology, Health Research Institute of the Jimenez Diaz Foundation, UAM, Madrid, Spain, UAM, Madrid, Spain
- Next Generation CART MAD Consortium, Madrid, Spain
| | - Beatriz Martin-Antonio
- Department of Experimental Hematology, Health Research Institute of the Jimenez Diaz Foundation, UAM, Madrid, Spain, UAM, Madrid, Spain
- Next Generation CART MAD Consortium, Madrid, Spain
- Departamento de Desarrollo de Medicamentos de Terapias Avanzadas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
11
|
Sima H, Shao W. Advancements in the design and function of bispecific CAR-T cells targeting B Cell-Associated tumor antigens. Int Immunopharmacol 2024; 142:113166. [PMID: 39298818 DOI: 10.1016/j.intimp.2024.113166] [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: 07/18/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
Single-targeted CAR-T has exhibited notable success in treating B-cell tumors, effectively improving patient outcomes. However, the recurrence rate among patients remains above fifty percent, primarily attributed to antigen escape and the diminished immune persistence of CAR-T cells. Over recent years, there has been a surge of interest in bispecific CAR-T cell therapies, marked by an increasing number of research articles and clinical applications annually. This paper undertakes a comprehensive review of influential studies on the design of bispecific CAR-T in recent years, examining their impact on bispecific CAR-T efficacy concerning disease classification, targeted antigens, and CAR design. Notable distinctions in antigen targeting within B-ALL, NHL, and MM are explored, along with an analysis of how CAR scFv, transmembrane region, hinge region, and co-stimulatory region design influence Bi-CAR-T efficacy across different tumors. The summary provided aims to serve as a reference for designing novel and improved CAR-Ts, facilitating more efficient treatment for B-cell malignant tumors.
Collapse
Affiliation(s)
- Helin Sima
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Wenwei Shao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China; Medical School of Tianjin University, Tianjin, China; State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin, China.
| |
Collapse
|
12
|
James SE, Chen S, Ng BD, Fischman JS, Jahn L, Boardman AP, Rajagopalan A, Elias HK, Massa A, Manuele D, Nichols KB, Lazrak A, Lee N, Roche AM, McFarland AG, Petrichenko A, Everett JK, Bushman FD, Fei T, Kousa AI, Lemarquis AL, DeWolf S, Peled JU, Vardhana SA, Klebanoff CA, van den Brink MRM. Leucine zipper-based immunomagnetic purification of CAR T cells displaying multiple receptors. Nat Biomed Eng 2024; 8:1592-1614. [PMID: 39715901 PMCID: PMC11917073 DOI: 10.1038/s41551-024-01287-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/26/2024] [Indexed: 12/25/2024]
Abstract
Resistance to chimaeric antigen receptor (CAR) T cell therapy develops through multiple mechanisms, most notably antigen loss and tumour-induced immune suppression. It has been suggested that T cells expressing multiple CARs may overcome the resistance of tumours and that T cells expressing receptors that switch inhibitory immune-checkpoint signals into costimulatory signals may enhance the activity of the T cells in the tumour microenvironment. However, engineering multiple features into a single T cell product is difficult because of the transgene-packaging constraints of current gene-delivery vectors. Here we describe a cell-sorting method that leverages leucine zippers for the selective single-step immunomagnetic purification of cells co-transduced with two vectors. Such 'Zip sorting' facilitated the generation of T cells simultaneously expressing up to four CARs and coexpressing up to three 'switch' receptors. In syngeneic mouse models, T cells with multiple CARs and multiple switch receptors eliminated antigenically heterogeneous populations of leukaemia cells coexpressing multiple inhibitory ligands. By combining diverse therapeutic strategies, Zip-sorted multi-CAR multi-switch-receptor T cells can overcome multiple mechanisms of CAR T cell resistance.
Collapse
Affiliation(s)
- Scott E James
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA.
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- City of Hope National Medical Center, Duarte, CA, USA.
| | - Sophia Chen
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- City of Hope National Medical Center, Duarte, CA, USA
| | - Brandon D Ng
- Weill Cornell Medical College, New York, NY, USA
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
| | - Jacob S Fischman
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Lorenz Jahn
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
| | - Alexander P Boardman
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Adhithi Rajagopalan
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- City of Hope National Medical Center, Duarte, CA, USA
| | - Harold K Elias
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alyssa Massa
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- City of Hope National Medical Center, Duarte, CA, USA
| | - Dylan Manuele
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
| | | | - Amina Lazrak
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
| | - Nicole Lee
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
| | - Aoife M Roche
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander G McFarland
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Angelina Petrichenko
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John K Everett
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Teng Fei
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anastasia I Kousa
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- City of Hope National Medical Center, Duarte, CA, USA
| | - Andri L Lemarquis
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- City of Hope National Medical Center, Duarte, CA, USA
| | - Susan DeWolf
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan U Peled
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Santosha A Vardhana
- Weill Cornell Medical College, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christopher A Klebanoff
- Weill Cornell Medical College, New York, NY, USA
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marcel R M van den Brink
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA.
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
13
|
Ai K, Liu B, Chen X, Huang C, Yang L, Zhang W, Weng J, Du X, Wu K, Lai P. Optimizing CAR-T cell therapy for solid tumors: current challenges and potential strategies. J Hematol Oncol 2024; 17:105. [PMID: 39501358 PMCID: PMC11539560 DOI: 10.1186/s13045-024-01625-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy demonstrates substantial efficacy in various hematological malignancies. However, its application in solid tumors is still limited. Clinical studies report suboptimal outcomes such as reduced cytotoxicity of CAR-T cells and tumor evasion, underscoring the need to address the challenges of sliding cytotoxicity in CAR-T cells. Despite improvements from fourth and next-generation CAR-T cells, new challenges include systemic toxicity from continuously secreted proteins, low productivity, and elevated costs. Recent research targets genetic modifications to boost killing potential, metabolic interventions to hinder tumor progression, and diverse combination strategies to enhance CAR-T cell therapy. Efforts to reduce the duration and cost of CAR-T cell therapy include developing allogenic and in-vivo approaches, promising significant future advancements. Concurrently, innovative technologies and platforms enhance the potential of CAR-T cell therapy to overcome limitations in treating solid tumors. This review explores strategies to optimize CAR-T cell therapies for solid tumors, focusing on enhancing cytotoxicity and overcoming application restrictions. We summarize recent advances in T cell subset selection, CAR-T structural modifications, infiltration enhancement, genetic and metabolic interventions, production optimization, and the integration of novel technologies, presenting therapeutic approaches that could improve CAR-T cell therapy's efficacy and applicability in solid tumors.
Collapse
Affiliation(s)
- Kexin Ai
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Bowen Liu
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China
| | - Xiaomei Chen
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China
| | - Chuxin Huang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Liping Yang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Weiya Zhang
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
| | - Jianyu Weng
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China
| | - Xin Du
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
- Cancer Center, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.
| | - Peilong Lai
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China.
| |
Collapse
|
14
|
Arunachalam AK, Grégoire C, Coutinho de Oliveira B, Melenhorst JJ. Advancing CAR T-cell therapies: Preclinical insights and clinical translation for hematological malignancies. Blood Rev 2024; 68:101241. [PMID: 39289094 DOI: 10.1016/j.blre.2024.101241] [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: 07/29/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has achieved significant success in achieving durable and potentially curative responses in patients with hematological malignancies. CARs are tailored fusion proteins that direct T cells to a specific antigen on tumor cells thereby eliciting a targeted immune response. The approval of several CD19-targeted CAR T-cell therapies has resulted in a notable surge in clinical trials involving CAR T cell therapies for hematological malignancies. Despite advancements in understanding response mechanisms, resistance patterns, and adverse events associated with CAR T-cell therapy, the translation of these insights into robust clinical efficacy has shown modest outcomes in both clinical trials and real-world scenarios. Therefore, the assessment of CAR T-cell functionality through rigorous preclinical studies plays a pivotal role in refining therapeutic strategies for clinical applications. This review provides an overview of the various in vitro and animal models used to assess the functionality of CAR T-cells. We discuss the findings from preclinical research involving approved CAR T-cell products, along with the implications derived from recent preclinical studies aiming to optimize the functionality of CAR T-cells. The review underscores the importance of robust preclinical evaluations and the need for models that accurately replicate human disease to bridge the gap between preclinical success and clinical efficacy.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Hematologic Neoplasms/therapy
- Hematologic Neoplasms/immunology
- Animals
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/transplantation
- Translational Research, Biomedical
- Disease Models, Animal
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
- Arun K Arunachalam
- Cell Therapy & Immuno-Engineering Program, Center for Immunotherapy and Precision Immuno-Oncology, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Céline Grégoire
- Cell Therapy & Immuno-Engineering Program, Center for Immunotherapy and Precision Immuno-Oncology, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Beatriz Coutinho de Oliveira
- Cell Therapy & Immuno-Engineering Program, Center for Immunotherapy and Precision Immuno-Oncology, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Jan Joseph Melenhorst
- Cell Therapy & Immuno-Engineering Program, Center for Immunotherapy and Precision Immuno-Oncology, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, United States of America.
| |
Collapse
|
15
|
Andrea AE, Chiron A, Sarrabayrouse G, Bessoles S, Hacein-Bey-Abina S. A structural, genetic and clinical comparison of CAR-T cells and CAR-NK cells: companions or competitors? Front Immunol 2024; 15:1459818. [PMID: 39430751 PMCID: PMC11486669 DOI: 10.3389/fimmu.2024.1459818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/16/2024] [Indexed: 10/22/2024] Open
Abstract
In recent years, following the groundbreaking achievements of chimeric antigen receptor (CAR) T cell therapy in hematological cancers, and advancements in cell engineering technologies, the exploration of other immune cells has garnered significant attention. CAR-Therapy extended beyond T cells to include CAR natural killer (NK) cells and CAR-macrophages, which are firmly established in the clinical trial landscape. Less conventional immune cells are also making their way into the scene, such as CAR mucosal-associated invariant T (MAIT) cells. This progress is advancing precision medicine and facilitating the development of ready-to-use biological treatments. However, in view of the unique features of natural killer cells, adoptive NK cell immunotherapy has emerged as a universal, allogenic, "off-the shelf" therapeutic strategy. CAR-NK cytotoxic cells present targeted tumor specificity but seem to be devoid of the side effects associated with CAR-T cells. CAR-NK cells appear to be potentially promising candidates for cancer immunotherapy. However, their application is hindered by significant challenges, particularly the limited persistence of CAR-NK cells in the body, which poses a hurdle to their sustained effectiveness in treating cancer. Based upon the foregoing, this review discusses the current status and applications of both CAR-T cells and CAR-NK cells in hematological cancers, and provides a comparative analysis of the structure, genetics, and clinical outcomes between these two types of genetically modified immune cells.
Collapse
Affiliation(s)
- Alain E. Andrea
- Department of Biology, Faculty of Arts and Sciences, Saint George University of Beirut, Beirut, Lebanon
| | - Andrada Chiron
- Université Paris Cité, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Unité des Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris Saclay, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France
| | - Guillaume Sarrabayrouse
- Université Paris Cité, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Unité des Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France
| | - Stéphanie Bessoles
- Université Paris Cité, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Unité des Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France
| | - Salima Hacein-Bey-Abina
- Université Paris Cité, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Unité des Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris Saclay, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France
| |
Collapse
|
16
|
Wu S, Luo Q, Li F, Zhang S, Zhang C, Liu J, Shao B, Hong Y, Tan T, Dong X, Chen B. Development of novel humanized CD19/BAFFR bicistronic chimeric antigen receptor T cells with potent antitumor activity against B-cell lineage neoplasms. Br J Haematol 2024; 205:1361-1373. [PMID: 38960449 DOI: 10.1111/bjh.19631] [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: 04/30/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024]
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy has shown remarkable efficacy in treating advanced B-cell malignancies by targeting CD19, but antigen-negative relapses and immune responses triggered by murine-derived antibodies remain significant challenges, necessitating the development of novel humanized multitarget CAR-T therapies. Here, we engineered a second-generation 4-1BB-CD3ζ-based CAR construct incorporating humanized CD19 single-chain variable fragments (scFvs) and BAFFR single-variable domains on heavy chains (VHHs), also known as nanobodies. The resultant CAR-T cells, with different constructs, were functionally compared both in vitro and in vivo. We found that the optimal tandem and bicistronic (BI) structures retained respective antigen-binding abilities, and both demonstrated specific activation when stimulated with target cells. At the same time, BI CAR-T cells (BI CARs) exhibited stronger tumour-killing ability and better secretion of interleukin-2 and tumour necrosis factor-alpha than single-target CAR-T cells. Additionally, BI CARs showed less exhaustion phenotype upon repeated antigen stimulation and demonstrated more potent and persistent antitumor effects in mouse xenograft models. Overall, we developed a novel humanized CD19/BAFFR bicistronic CAR (BI CAR) based on a combination of scFv and VHH, which showed potent and sustained antitumor ability both in vitro and in vivo, including against tumours with CD19 or BAFFR deficiencies.
Collapse
Affiliation(s)
- Sungui Wu
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Qian Luo
- Iaso Biotherapeutics Co. Ltd., Nanjing, Jiangsu, China
| | - Feiyu Li
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Suwen Zhang
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Cuiling Zhang
- Department of Hematology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jianwei Liu
- Iaso Biotherapeutics Co. Ltd., Nanjing, Jiangsu, China
| | - Bang Shao
- Iaso Biotherapeutics Co. Ltd., Nanjing, Jiangsu, China
| | - Yang Hong
- Department of Hematology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Taochao Tan
- Iaso Biotherapeutics Co. Ltd., Nanjing, Jiangsu, China
| | - Xiaoqing Dong
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Hematology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Bing Chen
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Hematology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
17
|
Lin H, Yang X, Ye S, Huang L, Mu W. Antigen escape in CAR-T cell therapy: Mechanisms and overcoming strategies. Biomed Pharmacother 2024; 178:117252. [PMID: 39098176 DOI: 10.1016/j.biopha.2024.117252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/06/2024] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has shown promise in treating hematological malignancies and certain solid tumors. However, its efficacy is often hindered by negative relapses resulting from antigen escape. This review firstly elucidates the mechanisms underlying antigen escape during CAR-T cell therapy, including the enrichment of pre-existing target-negative tumor clones, antigen gene mutations or alternative splicing, deficits in antigen processing, antigen redistribution, lineage switch, epitope masking, and trogocytosis-mediated antigen loss. Furthermore, we summarize various strategies to overcome antigen escape, evaluate their advantages and limitations, and propose future research directions. Thus, we aim to provide valuable insights to enhance the effectiveness of CAR-T cell therapy.
Collapse
Affiliation(s)
- Haolong Lin
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei 430030, China
| | - Xiuxiu Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei 430030, China
| | - Shanwei Ye
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei 430030, China
| | - Liang Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei 430030, China; State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600, China.
| | - Wei Mu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei 430030, China.
| |
Collapse
|
18
|
Spiga M, Martini E, Maffia MC, Ciceri F, Ruggiero E, Potenza A, Bonini C. Harnessing the tumor microenvironment to boost adoptive T cell therapy with engineered lymphocytes for solid tumors. Semin Immunopathol 2024; 46:8. [PMID: 39060547 DOI: 10.1007/s00281-024-01011-y] [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: 12/04/2023] [Accepted: 03/18/2024] [Indexed: 07/28/2024]
Abstract
Adoptive cell therapy (ACT) using Chimeric Antigen Receptor (CAR) and T Cell Receptor (TCR) engineered T cells represents an innovative therapeutic approach for the treatment of hematological malignancies, yet its application for solid tumors is still suboptimal. The tumor microenvironment (TME) places several challenges to overcome for a satisfactory therapeutic effect, such as physical barriers (fibrotic capsule and stroma), and inhibitory signals impeding T cell function. Some of these obstacles can be faced by combining ACT with other anti-tumor approaches, such as chemo/radiotherapy and checkpoint inhibitors. On the other hand, cutting edge technological tools offer the opportunity to overcome and, in some cases, take advantage of TME intrinsic characteristics to boost ACT efficacy. These include: the exploitation of chemokine gradients and integrin expression for preferential T-cell homing and extravasation; metabolic changes that have direct or indirect effects on TCR-T and CAR-T cells by increasing antigen presentation and reshaping T cell phenotype; introduction of additional synthetic receptors on TCR-T and CAR-T cells with the aim of increasing T cells survival and fitness.
Collapse
Affiliation(s)
- Martina Spiga
- Experimental Hematology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Martini
- Experimental Hematology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Chiara Maffia
- Experimental Hematology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Ciceri
- Vita-Salute San Raffaele University, Milan, Italy
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Eliana Ruggiero
- Experimental Hematology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessia Potenza
- Experimental Hematology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Chiara Bonini
- Experimental Hematology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
19
|
Wang X, Zhang C, Su J, Ren S, Wang X, Zhang Y, Yuan Z, He X, Wu X, Li M, Du F, Chen Y, Deng S, Zhao Y, Wang X, Sun Y, Shen J, Ji H, Hou Y, Xiao Z. Rejuvenation Strategy for Inducing and Enhancing Autoimmune Response to Eliminate Senescent Cells. Aging Dis 2024:AD.2024.0579. [PMID: 39122450 DOI: 10.14336/ad.2024.0579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
The process of aging, which involves progressive changes in the body over time, is closely associated with the development of age-related diseases. Cellular senescence is a pivotal hallmark and mechanism of the aging process. The accumulation of senescent cells can significantly contribute to the onset of age-related diseases, thereby compromising overall health. Conversely, the elimination of senescent cells enhances the body's regenerative and reparative capacity, thereby retarding the aging process. Here, we present a brief overview of 12 Hallmarks of aging and subsequently emphasize the potential of immune checkpoint blockade, innate immune cell therapy (including T cells, iNKT cells, macrophages, and NK cells), as well as CAR-T cell therapy for inducing and augmenting immune responses aimed at eliminating senescent cells. In addition to CAR-T cells, we also explore the possibility of engineered immune cells such as CAR-NK and CAR-M cells to eliminate senescent cells. In summary, immunotherapy, as an emerging strategy for the treatment of aging, offers new prospects for age-related research.
Collapse
Affiliation(s)
- Xingyue Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Chengyu Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jiahong Su
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Siqi Ren
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yinping Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Zijun Yuan
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xinyu He
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Shuai Deng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Xiaodong Wang
- Department of Hepatobiliary Disease, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Huijiao Ji
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yunqing Hou
- LongmaTan District People's Hospital of Luzhou City, Luzhou 646600, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
- Department of Pharmacology, School of Pharmacy, Sichuan College of Traditional Chinese Medicine, Mianyang 621000, China
| |
Collapse
|
20
|
Ng BD, Rajagopalan A, Kousa AI, Fischman JS, Chen S, Massa A, Elias HK, Manuele D, Galiano M, Lemarquis AL, Boardman AP, DeWolf S, Pierce J, Bogen B, James SE, van den Brink MRM. IL-18-secreting multiantigen targeting CAR T cells eliminate antigen-low myeloma in an immunocompetent mouse model. Blood 2024; 144:171-186. [PMID: 38579288 PMCID: PMC11302468 DOI: 10.1182/blood.2023022293] [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: 09/15/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/07/2024] Open
Abstract
ABSTRACT Multiple myeloma is a plasma cell malignancy that is currently incurable with conventional therapies. Following the success of CD19-targeted chimeric antigen receptor (CAR) T cells in leukemia and lymphoma, CAR T cells targeting B-cell maturation antigen (BCMA) more recently demonstrated impressive activity in relapsed and refractory myeloma patients. However, BCMA-directed therapy can fail due to weak expression of BCMA on myeloma cells, suggesting that novel approaches to better address this antigen-low disease may improve patient outcomes. We hypothesized that engineered secretion of the proinflammatory cytokine interleukin-18 (IL-18) and multiantigen targeting could improve CAR T-cell activity against BCMA-low myeloma. In a syngeneic murine model of myeloma, CAR T cells targeting the myeloma-associated antigens BCMA and B-cell activating factor receptor (BAFF-R) failed to eliminate myeloma when these antigens were weakly expressed, whereas IL-18-secreting CAR T cells targeting these antigens promoted myeloma clearance. IL-18-secreting CAR T cells developed an effector-like T-cell phenotype, promoted interferon-gamma production, reprogrammed the myeloma bone marrow microenvironment through type-I/II interferon signaling, and activated macrophages to mediate antimyeloma activity. Simultaneous targeting of weakly-expressed BCMA and BAFF-R with dual-CAR T cells enhanced T-cell:target-cell avidity, increased overall CAR signal strength, and stimulated antimyeloma activity. Dual-antigen targeting augmented CAR T-cell secretion of engineered IL-18 and facilitated elimination of larger myeloma burdens in vivo. Our results demonstrate that combination of engineered IL-18 secretion and multiantigen targeting can eliminate myeloma with weak antigen expression through distinct mechanisms.
Collapse
Affiliation(s)
- Brandon D. Ng
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pharmacology, Weill Cornell Medicine, New York, NY
| | - Adhithi Rajagopalan
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Anastasia I. Kousa
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Jacob S. Fischman
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA
| | - Sophia Chen
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alyssa Massa
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Harold K. Elias
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Dylan Manuele
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Michael Galiano
- Molecular Cytology Core, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andri L. Lemarquis
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alexander P. Boardman
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Susan DeWolf
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jonah Pierce
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Immunology and Microbial Pathogenesis, Weill Cornell Medicine, New York, NY
| | | | - Scott E. James
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY
- Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marcel R. M. van den Brink
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- City of Hope Comprehensive Cancer Center, Duarte, CA
- Department of Immunology and Microbial Pathogenesis, Weill Cornell Medicine, New York, NY
- Department of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY
- Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
21
|
Van der Vreken A, Vanderkerken K, De Bruyne E, De Veirman K, Breckpot K, Menu E. Fueling CARs: metabolic strategies to enhance CAR T-cell therapy. Exp Hematol Oncol 2024; 13:66. [PMID: 38987856 PMCID: PMC11238373 DOI: 10.1186/s40164-024-00535-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024] Open
Abstract
CAR T cells are widely applied for relapsed hematological cancer patients. With six approved cell therapies, for Multiple Myeloma and other B-cell malignancies, new insights emerge. Profound evidence shows that patients who fail CAR T-cell therapy have, aside from antigen escape, a more glycolytic and weakened metabolism in their CAR T cells, accompanied by a short lifespan. Recent advances show that CAR T cells can be metabolically engineered towards oxidative phosphorylation, which increases their longevity via epigenetic and phenotypical changes. In this review we elucidate various strategies to rewire their metabolism, including the design of the CAR construct, co-stimulus choice, genetic modifications of metabolic genes, and pharmacological interventions. We discuss their potential to enhance CAR T-cell functioning and persistence through memory imprinting, thereby improving outcomes. Furthermore, we link the pharmacological treatments with their anti-cancer properties in hematological malignancies to ultimately suggest novel combination strategies.
Collapse
Affiliation(s)
- Arne Van der Vreken
- Translational Oncology Research Center, Team Hematology and Immunology, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Karin Vanderkerken
- Translational Oncology Research Center, Team Hematology and Immunology, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Elke De Bruyne
- Translational Oncology Research Center, Team Hematology and Immunology, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Kim De Veirman
- Translational Oncology Research Center, Team Hematology and Immunology, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Karine Breckpot
- Translational Oncology Research Center, Team Laboratory of Cellular and Molecular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Eline Menu
- Translational Oncology Research Center, Team Hematology and Immunology, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium.
| |
Collapse
|
22
|
Lakhani A, Chen X, Chen LC, Hong M, Khericha M, Chen Y, Chen YY, Park JO. Extracellular domains of CARs reprogramme T cell metabolism without antigen stimulation. Nat Metab 2024; 6:1143-1160. [PMID: 38658805 PMCID: PMC11845092 DOI: 10.1038/s42255-024-01034-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 03/25/2024] [Indexed: 04/26/2024]
Abstract
Metabolism is an indispensable part of T cell proliferation, activation and exhaustion, yet the metabolism of chimeric antigen receptor (CAR)-T cells remains incompletely understood. CARs are composed of extracellular domains-often single-chain variable fragments (scFvs)-that determine ligand specificity and intracellular domains that trigger signalling following antigen binding. Here, we show that CARs differing only in the scFv variously reprogramme T cell metabolism. Even without exposure to antigens, some CARs increase proliferation and nutrient uptake in T cells. Using stable isotope tracers and mass spectrometry, we observed basal metabolic fluxes through glycolysis doubling and amino acid uptake overtaking anaplerosis in CAR-T cells harbouring a rituximab scFv, unlike other similar anti-CD20 scFvs. Disparate rituximab and 14G2a-based anti-GD2 CAR-T cells are similarly hypermetabolic and channel excess nutrients to nitrogen overflow metabolism. Modest overflow metabolism of CAR-T cells and metabolic compatibility between cancer cells and CAR-T cells are identified as features of efficacious CAR-T cell therapy.
Collapse
Affiliation(s)
- Aliya Lakhani
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ximin Chen
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Laurence C Chen
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Mihe Hong
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Mobina Khericha
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yu Chen
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yvonne Y Chen
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, CA, USA
- Parker Institute for Cancer Immunotherapy at UCLA, Los Angeles, CA, USA
| | - Junyoung O Park
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
23
|
Lin HK, Uricoli B, Freeman RM, Hossian AKMN, He Z, Anderson JYL, Neffling M, Legier JM, Blake DA, Doxie DB, Nair R, Koff JL, Dhodapkar KM, Shanmugam M, Dreaden EC, Rafiq S. Engineering Improved CAR T Cell Products with A Multi-Cytokine Particle Platform for Hematologic and Solid Tumors. Adv Healthc Mater 2024; 13:e2302425. [PMID: 38245855 PMCID: PMC11144092 DOI: 10.1002/adhm.202302425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/05/2024] [Indexed: 01/22/2024]
Abstract
Despite the remarkable clinical efficacy of chimeric antigen receptor (CAR) T cells in hematological malignancies, only a subset of patients achieves a durable complete response (dCR). DCR has been correlated with CAR T cell products enriched with T cells memory phenotypes. Therefore, reagents that consistently promote memory phenotypes during the manufacturing of CAR T cells have the potential to significantly improve clinical outcomes. A novel modular multi-cytokine particle (MCP) platform is developed that combines the signals necessary for activation, costimulation, and cytokine support into a single "all-in-one" stimulation reagent for CAR T cell manufacturing. This platform allows for the assembly and screening of compositionally diverse MCP libraries to identify formulations tailored to promote specific phenotypes with a high degree of flexibility. The approach is leveraged to identify unique MCP formulations that manufacture CAR T cell products from diffuse large B cell patients with increased proportions of memory-like phenotypes MCP-manufactured CAR T cells demonstrate superior anti-tumor efficacy in mouse models of lymphoma and ovarian cancer through enhanced persistence. These findings serve as a proof-of-principle of the powerful utility of the MCP platform to identify "all-in-one" stimulation reagents that can improve the effectiveness of cell therapy products through optimal manufacturing.
Collapse
Affiliation(s)
- Heather K. Lin
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Biaggio Uricoli
- Wallace H. Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology Atlanta, GA, USA
| | - Ruby M. Freeman
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - AKM Nawshad Hossian
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Zhulin He
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | - Jonathan M. Legier
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Dejah A. Blake
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Deon B. Doxie
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Atlanta, GA, USA
| | - Remya Nair
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jean L. Koff
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Atlanta, GA, USA
| | - Kavita M. Dhodapkar
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA, USA
| | - Mala Shanmugam
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Atlanta, GA, USA
| | - Erik C. Dreaden
- Wallace H. Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Atlanta, GA, USA
| | - Sarwish Rafiq
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Atlanta, GA, USA
| |
Collapse
|
24
|
Brillembourg H, Martínez-Cibrián N, Bachiller M, Alserawan L, Ortiz-Maldonado V, Guedan S, Delgado J. The role of chimeric antigen receptor T cells targeting more than one antigen in the treatment of B-cell malignancies. Br J Haematol 2024; 204:1649-1659. [PMID: 38362778 DOI: 10.1111/bjh.19348] [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: 11/11/2023] [Revised: 01/24/2024] [Accepted: 02/07/2024] [Indexed: 02/17/2024]
Abstract
Several products containing chimeric antigen receptor T cells targeting CD19 (CART19) have been approved for the treatment of patients with relapsed/refractory non-Hodgkin's lymphoma (NHL) and acute lymphoblastic leukaemia (ALL). Despite very impressive response rates, a significant percentage of patients experience disease relapse and die of progressive disease. A major cause of CART19 failure is loss or downregulation of CD19 expression in tumour cells, which has prompted a myriad of novel strategies aimed at targeting more than one antigen (e.g. CD19 and CD20 or CD22). Dual targeting can the accomplished through co-administration of two separate products, co-transduction with two different vectors, bicistronic cassettes or tandem receptors. In this manuscript, we review the pros and cons of each strategy and the clinical results obtained so far.
Collapse
Affiliation(s)
| | - Núria Martínez-Cibrián
- Department of Haematology, Hospital Clínic, Barcelona, Spain
- Oncology and Haematology Area, FRCB-IDIBAPS, Barcelona, Spain
| | - Mireia Bachiller
- Oncology and Haematology Area, FRCB-IDIBAPS, Barcelona, Spain
- Department of Clinical Pharmacology, Hospital Clínic, Barcelona, Spain
| | | | - Valentín Ortiz-Maldonado
- Department of Haematology, Hospital Clínic, Barcelona, Spain
- Oncology and Haematology Area, FRCB-IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Sònia Guedan
- Oncology and Haematology Area, FRCB-IDIBAPS, Barcelona, Spain
| | - Julio Delgado
- Department of Haematology, Hospital Clínic, Barcelona, Spain
- Oncology and Haematology Area, FRCB-IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
- CIBERONC, Madrid, Spain
| |
Collapse
|
25
|
Khaniya A, Rad SMAH, Halpin J, Tawinwung S, McLellan A, Suppipat K, Hirankarn N. Development of a compact bidirectional promoter-driven dual chimeric antigen receptor (CAR) construct targeting CD19 and CD20 in the Sleeping Beauty (SB) transposon system. J Immunother Cancer 2024; 12:e008555. [PMID: 38677881 PMCID: PMC11057265 DOI: 10.1136/jitc-2023-008555] [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] [Accepted: 04/11/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND A bidirectional promoter-driven chimeric antigen receptor (CAR) cassette provides the simultaneous expression of two CARs, which significantly enhances dual antigen-targeted CAR T-cell therapy. METHODS We developed a second-generation CAR directing CD19 and CD20 antigens, incorporating them in a head-to-head orientation from a bidirectional promoter using a single Sleeping Beauty transposon system. The efficacy of bidirectional promoter-driven dual CD19 and CD20 CAR T cells was determined in vitro against cell lines expressing either, or both, CD19 and CD20 antigens. In vivo antitumor activity was tested in Raji lymphoma-bearing immunodeficient NOD-scid IL2Rgammanull (NSG) mice. RESULTS Of all tested promoters, the bidirectional EF-1α promoter optimally expressed transcripts from both sense (CD19-CAR) and antisense (GFP.CD20-CAR) directions. Superior cytotoxicity, cytokine production and antigen-specific activation were observed in vitro in the bidirectional EF-1α promoter-driven CD19/CD20 CAR T cells. In contrast, a unidirectional construct driven by the EF-1α promoter, but using self-cleaving peptide-linked CD19 and CD20 CARs, showed inferior expression and in vitro function. Treatment of mice bearing advanced Raji lymphomas with bidirectional EF-1α promoter-driven CD19/CD20 CAR T cells effectively controlled tumor growth and extended the survival of mice compared with group treated with single antigen targeted CAR T cells. CONCLUSION The use of bidirectional promoters in a single vector offers advantages of size and robust CAR expression with the potential to expand use in other forms of gene therapies like CAR T cells.
Collapse
MESH Headings
- Antigens, CD19/immunology
- Antigens, CD19/genetics
- Humans
- Animals
- Antigens, CD20/genetics
- Antigens, CD20/metabolism
- Antigens, CD20/immunology
- Mice
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/immunology
- DNA Transposable Elements
- Promoter Regions, Genetic
- Immunotherapy, Adoptive/methods
- Mice, Inbred NOD
- Cell Line, Tumor
- Mice, SCID
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Asmita Khaniya
- Medical Microbiology, Chulalongkorn University, Bangkok, Thailand
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand
| | | | - Josh Halpin
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Supannikar Tawinwung
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand
- Pharmacology and Physiology, Chulalongkorn University Faculty of Pharmaceutical Sciences, Bangkok, Thailand
| | - Alexander McLellan
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Koramit Suppipat
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Research Affairs, Chulalongkorn University, Bangkok, Thailand
| | - Nattiya Hirankarn
- Center of Excellence in Immunology and Immune-mediated Diseases, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
26
|
Amorós-Pérez B, Rivas-Pardo B, Gómez del Moral M, Subiza JL, Martínez-Naves E. State of the Art in CAR-T Cell Therapy for Solid Tumors: Is There a Sweeter Future? Cells 2024; 13:725. [PMID: 38727261 PMCID: PMC11083689 DOI: 10.3390/cells13090725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 05/13/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has proven to be a powerful treatment for hematological malignancies. The situation is very different in the case of solid tumors, for which no CAR-T-based therapy has yet been approved. There are many factors contributing to the absence of response in solid tumors to CAR-T cells, such as the immunosuppressive tumor microenvironment (TME), T cell exhaustion, or the lack of suitable antigen targets, which should have a stable and specific expression on tumor cells. Strategies being developed to improve CAR-T-based therapy for solid tumors include the use of new-generation CARs such as TRUCKs or bi-specific CARs, the combination of CAR therapy with chemo- or radiotherapy, the use of checkpoint inhibitors, and the use of oncolytic viruses. Furthermore, despite the scarcity of targets, a growing number of phase I/II clinical trials are exploring new solid-tumor-associated antigens. Most of these antigens are of a protein nature; however, there is a clear potential in identifying carbohydrate-type antigens associated with tumors, or carbohydrate and proteoglycan antigens that emerge because of aberrant glycosylations occurring in the context of tumor transformation.
Collapse
Affiliation(s)
- Beatriz Amorós-Pérez
- Department of Immunology, Ophthalmology and ORL, School of Medicine, Universidad Complutense of Madrid (UCM), 28040 Madrid, Spain; (B.A.-P.); (B.R.-P.)
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Inmunotek S.L., 28805 Madrid, Spain;
| | - Benigno Rivas-Pardo
- Department of Immunology, Ophthalmology and ORL, School of Medicine, Universidad Complutense of Madrid (UCM), 28040 Madrid, Spain; (B.A.-P.); (B.R.-P.)
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Manuel Gómez del Moral
- Department of Cellular Biology, School of Medicine, Universidad Complutense of Madrid (UCM), 28040 Madrid, Spain;
| | | | - Eduardo Martínez-Naves
- Department of Immunology, Ophthalmology and ORL, School of Medicine, Universidad Complutense of Madrid (UCM), 28040 Madrid, Spain; (B.A.-P.); (B.R.-P.)
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| |
Collapse
|
27
|
Pang Y, Ghosh N. Novel and multiple targets for chimeric antigen receptor-based therapies in lymphoma. Front Oncol 2024; 14:1396395. [PMID: 38711850 PMCID: PMC11070555 DOI: 10.3389/fonc.2024.1396395] [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: 03/05/2024] [Accepted: 04/08/2024] [Indexed: 05/08/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy targeting CD19 in B-cell non-Hodgkin lymphoma (NHL) validates the utility of CAR-based therapy for lymphomatous malignancies. Despite the success, treatment failure due to CD19 antigen loss, mutation, or down-regulation remains the main obstacle to cure. On-target, off-tumor effect of CD19-CAR T leads to side effects such as prolonged B-cell aplasia, limiting the application of therapy in indolent diseases such as chronic lymphocytic leukemia (CLL). Alternative CAR targets and multi-specific CAR are potential solutions to improving cellular therapy outcomes in B-NHL. For Hodgkin lymphoma and T-cell lymphoma, several cell surface antigens have been studied as CAR targets, some of which already showed promising results in clinical trials. Some antigens are expressed by different lymphomas and could be used for designing tumor-agnostic CAR. Here, we reviewed the antigens that have been studied for novel CAR-based therapies, as well as CARs designed to target two or more antigens in the treatment of lymphoma.
Collapse
Affiliation(s)
- Yifan Pang
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Wake Forest School of Medicine, Charlotte, NC, United States
| | | |
Collapse
|
28
|
Dabkowska A, Domka K, Firczuk M. Advancements in cancer immunotherapies targeting CD20: from pioneering monoclonal antibodies to chimeric antigen receptor-modified T cells. Front Immunol 2024; 15:1363102. [PMID: 38638442 PMCID: PMC11024268 DOI: 10.3389/fimmu.2024.1363102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/25/2024] [Indexed: 04/20/2024] Open
Abstract
CD20 located predominantly on the B cells plays a crucial role in their development, differentiation, and activation, and serves as a key therapeutic target for the treatment of B-cell malignancies. The breakthrough of monoclonal antibodies directed against CD20, notably exemplified by rituximab, revolutionized the prognosis of B-cell malignancies. Rituximab, approved across various hematological malignancies, marked a paradigm shift in cancer treatment. In the current landscape, immunotherapies targeting CD20 continue to evolve rapidly. Beyond traditional mAbs, advancements include antibody-drug conjugates (ADCs), bispecific antibodies (BsAbs), and chimeric antigen receptor-modified (CAR) T cells. ADCs combine the precision of antibodies with the cytotoxic potential of drugs, presenting a promising avenue for enhanced therapeutic efficacy. BsAbs, particularly CD20xCD3 constructs, redirect cytotoxic T cells to eliminate cancer cells, thereby enhancing both precision and potency in their therapeutic action. CAR-T cells stand as a promising strategy for combatting hematological malignancies, representing one of the truly personalized therapeutic interventions. Many new therapies are currently being evaluated in clinical trials. This review serves as a comprehensive summary of CD20-targeted therapies, highlighting the progress and challenges that persist. Despite significant advancements, adverse events associated with these therapies and the development of resistance remain critical issues. Understanding and mitigating these challenges is paramount for the continued success of CD20-targeted immunotherapies.
Collapse
Affiliation(s)
- Agnieszka Dabkowska
- Laboratory of Immunology, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Krzysztof Domka
- Laboratory of Immunology, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Malgorzata Firczuk
- Laboratory of Immunology, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
29
|
Ayala Ceja M, Khericha M, Harris CM, Puig-Saus C, Chen YY. CAR-T cell manufacturing: Major process parameters and next-generation strategies. J Exp Med 2024; 221:e20230903. [PMID: 38226974 PMCID: PMC10791545 DOI: 10.1084/jem.20230903] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/02/2023] [Accepted: 12/14/2023] [Indexed: 01/17/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapies have demonstrated strong curative potential and become a critical component in the array of B-cell malignancy treatments. Successful deployment of CAR-T cell therapies to treat hematologic and solid cancers, as well as other indications such as autoimmune diseases, is dependent on effective CAR-T cell manufacturing that impacts not only product safety and efficacy but also overall accessibility to patients in need. In this review, we discuss the major process parameters of autologous CAR-T cell manufacturing, as well as regulatory considerations and ongoing developments that will enable the next generation of CAR-T cell therapies.
Collapse
Affiliation(s)
- Melanie Ayala Ceja
- Department of Microbiology, Immunology, and Molecular Genetics, University of California−Los Angeles, Los Angeles, CA, USA
| | - Mobina Khericha
- Department of Microbiology, Immunology, and Molecular Genetics, University of California−Los Angeles, Los Angeles, CA, USA
| | - Caitlin M. Harris
- Department of Microbiology, Immunology, and Molecular Genetics, University of California−Los Angeles, Los Angeles, CA, USA
| | - Cristina Puig-Saus
- Department of Medicine, University of California−Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California−Los Angeles, Los Angeles, CA, USA
- Parker Institute for Cancer Immunotherapy Center at University of California−Los Angeles, Los Angeles, CA, USA
| | - Yvonne Y. Chen
- Department of Microbiology, Immunology, and Molecular Genetics, University of California−Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California−Los Angeles, Los Angeles, CA, USA
- Parker Institute for Cancer Immunotherapy Center at University of California−Los Angeles, Los Angeles, CA, USA
- Department of Chemical and Biomolecular Engineering, University of California−Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
30
|
Testa U, D’Alò F, Pelosi E, Castelli G, Leone G. CAR-T Cell Therapy for Follicular Lymphomas. Mediterr J Hematol Infect Dis 2024; 16:e2024012. [PMID: 38223488 PMCID: PMC10786124 DOI: 10.4084/mjhid.2024.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 12/15/2023] [Indexed: 01/16/2024] Open
Abstract
Follicular lymphoma is the second most diagnosed lymphoma in Western Europe. Significant advancements have considerably improved the survival of FL patients. However, 10-20% of these patients are refractory to standard treatments, and most of them will relapse. The treatment of follicular lymphoma patients with multiply relapsed or refractory disease represents an area of high-unmet needing new treatments with stronger efficacy. Chimeric antigen receptor (CAR)-T cell therapy targeting B-cell antigens, such as CD19 or CD20, is emerging as an efficacious treatment for R/R follicular lymphoma patients, particularly for those with early relapse and refractory to alkylating agents and to anti-CD20 monoclonal antibodies, resulting in a high rate of durable responses in a high proportion of patients.
Collapse
Affiliation(s)
| | - Francesco D’Alò
- Dipartimento Di Diagnostica per Immagini, Radioterapia Oncologica Ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy. Sezione Di Ematologia
- Dipartimento Di Scienze Radiologiche Ed Ematologiche, Università Cattolica Del Sacro Cuore, Roma, Italy
| | | | | | - Giuseppe Leone
- Dipartimento Di Scienze Radiologiche Ed Ematologiche, Università Cattolica Del Sacro Cuore, Roma, Italy
| |
Collapse
|
31
|
Ruella M, Korell F, Porazzi P, Maus MV. Mechanisms of resistance to chimeric antigen receptor-T cells in haematological malignancies. Nat Rev Drug Discov 2023; 22:976-995. [PMID: 37907724 PMCID: PMC10965011 DOI: 10.1038/s41573-023-00807-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 11/02/2023]
Abstract
Chimeric antigen receptor (CAR)-T cells have recently emerged as a powerful therapeutic approach for the treatment of patients with chemotherapy-refractory or relapsed blood cancers, including acute lymphoblastic leukaemia, diffuse large B cell lymphoma, follicular lymphoma, mantle cell lymphoma and multiple myeloma. Nevertheless, resistance to CAR-T cell therapies occurs in most patients. In this Review, we summarize the resistance mechanisms to CAR-T cell immunotherapy by analysing CAR-T cell dysfunction, intrinsic tumour resistance and the immunosuppressive tumour microenvironment. We discuss current research strategies to overcome multiple resistance mechanisms, including optimization of the CAR design, improvement of in vivo T cell function and persistence, modulation of the immunosuppressive tumour microenvironment and synergistic combination strategies.
Collapse
Affiliation(s)
- Marco Ruella
- Division of Hematology and Oncology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Felix Korell
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Patrizia Porazzi
- Division of Hematology and Oncology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
32
|
Chu F, Cao J, Liu J, Yang H, Davis TJ, Kuang SQ, Cheng X, Zhang Z, Karri S, Vien LT, Bover L, Sun R, Vega F, Green M, Davis RE, Neelapu SS. Chimeric antigen receptor T cells to target CD79b in B-cell lymphomas. J Immunother Cancer 2023; 11:e007515. [PMID: 38007239 PMCID: PMC10680003 DOI: 10.1136/jitc-2023-007515] [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] [Accepted: 10/17/2023] [Indexed: 11/27/2023] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T cells targeting CD19 mediate potent and durable effects in B-cell malignancies. However, antigen loss or downregulation is a frequent cause of resistance. Here, we report development of a novel CAR T-cell therapy product to target CD79b, a pan B-cell antigen, widely expressed in most B-cell lymphomas. METHODS We generated a novel anti-CD79b monoclonal antibody by hybridoma method. The specificity of the antibody was determined by testing against isogenic cell lines with human CD79b knock-in or knock-out. A single-chain variable fragment derived from the monoclonal antibody was used to make a panel of CD79b-targeting CAR molecules containing various hinge, transmembrane, and co-stimulatory domains. These were lentivirally transduced into primary T cells and tested for antitumor activity in in vitro and in vivo B-cell lymphoma models. RESULTS We found that the novel anti-CD79b monoclonal antibody was highly specific and bound only to human CD79b and no other cell surface protein. In testing the various CD79b-targeting CAR molecules, superior antitumor efficacy in vitro and in vivo was found for a CAR consisting CD8α hinge and transmembrane domains, an OX40 co-stimulatory domain, and a CD3ζ signaling domain. This CD79b CAR specifically recognized human CD79b-expressing lymphoma cell lines but not CD79b knock-out cell lines. CD79b CAR T cells, generated from T cells from either healthy donors or patients with lymphoma, proliferated, produced cytokines, degranulated, and exhibited robust cytotoxic activity in vitro against CD19+ and CD19- lymphoma cell lines and patient-derived lymphoma tumors relapsing after prior CD19 CAR T-cell therapy. Furthermore, CD79b CAR T cells were highly efficient at eradicating pre-established lymphoma tumors in vivo in three aggressive lymphoma xenograft models, including two cell line-derived xenografts and one patient-derived xenograft. Notably, these CAR T cells did not demonstrate any significant tonic signaling activity or markers of exhaustion. CONCLUSION Our results indicated that this novel CD79b CAR T-cell therapy product has robust antitumor activity against B-cell lymphomas. These results supported initiation of a phase 1 clinical trial to evaluate this product in patients with relapsed or refractory B-cell lymphomas.
Collapse
Affiliation(s)
- Fuliang Chu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jingjing Cao
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jingwei Liu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Haopeng Yang
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Timothy J Davis
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shao-Qing Kuang
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xiaoyun Cheng
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zheng Zhang
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Swathi Karri
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Long T Vien
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Laura Bover
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ryan Sun
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Francisco Vega
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael Green
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Richard Eric Davis
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sattva S Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
33
|
Stock S, Klüver AK, Fertig L, Menkhoff VD, Subklewe M, Endres S, Kobold S. Mechanisms and strategies for safe chimeric antigen receptor T-cell activity control. Int J Cancer 2023; 153:1706-1725. [PMID: 37350095 DOI: 10.1002/ijc.34635] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 05/07/2023] [Accepted: 06/02/2023] [Indexed: 06/24/2023]
Abstract
The clinical application of chimeric antigen receptor (CAR) T-cell therapy has rapidly changed the treatment options for terminally ill patients with defined blood-borne cancer types. However, CAR T-cell therapy can lead to severe therapy-associated toxicities including CAR-related hematotoxicity, ON-target OFF-tumor toxicity, cytokine release syndrome (CRS) or immune effector cell-associated neurotoxicity syndrome (ICANS). Just as CAR T-cell therapy has evolved regarding receptor design, gene transfer systems and production protocols, the management of side effects has also improved. However, because of measures taken to abrogate adverse events, CAR T-cell viability and persistence might be impaired before complete remission can be achieved. This has fueled efforts for the development of extrinsic and intrinsic strategies for better control of CAR T-cell activity. These approaches can mediate a reversible resting state or irreversible T-cell elimination, depending on the route chosen. Control can be passive or active. By combination of CAR T-cells with T-cell inhibiting compounds, pharmacologic control, mostly independent of the CAR construct design used, can be achieved. Other strategies involve the genetic modification of T-cells or further development of the CAR construct by integration of molecular ON/OFF switches such as suicide genes. Alternatively, CAR T-cell activity can be regulated intracellularly through a self-regulation function or extracellularly through titration of a CAR adaptor or of a priming small molecule. In this work, we review the current strategies and mechanisms to control activity of CAR T-cells reversibly or irreversibly for preventing and for managing therapy-associated toxicities.
Collapse
Affiliation(s)
- Sophia Stock
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
- Department of Medicine III, LMU University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Anna-Kristina Klüver
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Luisa Fertig
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Vivien D Menkhoff
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Marion Subklewe
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Stefan Endres
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany
| |
Collapse
|
34
|
Hoces D, Miguens Blanco J, Hernández-López RA. A synthetic biology approach to engineering circuits in immune cells. Immunol Rev 2023; 320:120-137. [PMID: 37464881 DOI: 10.1111/imr.13244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 06/08/2023] [Indexed: 07/20/2023]
Abstract
A synthetic circuit in a biological system involves the designed assembly of genetic elements, biomolecules, or cells to create a defined function. These circuits are central in synthetic biology, enabling the reprogramming of cellular behavior and the engineering of cells with customized responses. In cancer therapeutics, engineering T cells with circuits have the potential to overcome the challenges of current approaches, for example, by allowing specific recognition and killing of cancer cells. Recent advances also facilitate engineering integrated circuits for the controlled release of therapeutic molecules at specified locations, for example, in a solid tumor. In this review, we discuss recent strategies and applications of synthetic receptor circuits aimed at enhancing immune cell functions for cancer immunotherapy. We begin by introducing the concept of circuits in networks at the molecular and cellular scales and provide an analysis of the development and implementation of several synthetic circuits in T cells that have the goal to overcome current challenges in cancer immunotherapy. These include specific targeting of cancer cells, increased T-cell proliferation, and persistence in the tumor microenvironment. By harnessing the power of synthetic biology, and the characteristics of certain circuit architectures, it is now possible to engineer a new generation of immune cells that recognize cancer cells, while minimizing off-target toxicities. We specifically discuss T-cell circuits for antigen density sensing. These circuits allow targeting of solid tumors that share antigens with normal tissues. Additionally, we explore designs for synthetic circuits that could control T-cell differentiation or T-cell fate as well as the concept of synthetic multicellular circuits that leverage cellular communication and division of labor to achieve improved therapeutic efficacy. As our understanding of cell biology expands and novel tools for genome, protein, and cell engineering are developed, we anticipate further innovative approaches to emerge in the design and engineering of circuits in immune cells.
Collapse
Affiliation(s)
- Daniel Hoces
- Department of Bioengineering, Stanford University, Stanford, California, USA
- Department of Genetics, Stanford University, Stanford, California, USA
| | - Jesús Miguens Blanco
- Department of Bioengineering, Stanford University, Stanford, California, USA
- Department of Genetics, Stanford University, Stanford, California, USA
| | - Rogelio A Hernández-López
- Department of Bioengineering, Stanford University, Stanford, California, USA
- Department of Genetics, Stanford University, Stanford, California, USA
- Stanford Cancer Institute, Stanford, California, USA
- Chan-Zuckerberg Biohub-San Francisco, San Francisco, California, USA
| |
Collapse
|
35
|
Rodríguez Gil de Montes AL, Spencer LM. Chimeric Antigen Receptor T Cells: Immunotherapy for the Treatment of Leukemia, Lymphoma, and Myeloma. Mol Cancer Ther 2023; 22:1261-1269. [PMID: 37596239 DOI: 10.1158/1535-7163.mct-23-0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 05/27/2023] [Accepted: 08/15/2023] [Indexed: 08/20/2023]
Abstract
In immunotherapy with T cells genetically modified to express chimeric antigen receptors (CAR), autologous lymphocytes are extracted from the patient, genetically modified to obtain CAR-T cells, and reintroduced into the patient to attack cancer cells. The success of this therapy has been achieved in the area of CD19-positive leukemias and lymphomas, being approved for the treatment of non-Hodgkin's lymphomas, acute lymphoblastic leukemia, and multiple myeloma. CARs are proteins that combine antibody specificity with T-cell cytotoxicity. The most common toxicities associated with therapy were not predicted by preclinical testing and include cytokine release syndrome, neurotoxicity, and cytopenias. These toxicities are usually reversible. One of the main challenges facing the field is the high economic cost that therapy entails, so the search for ways to reduce this cost must be a priority. In addition, other challenges to overcome include the situation that not all patients are supplied with the product and the existence of long waiting times for the start of therapy. The aim of this review is to present the development of the structure of CAR-T cells, the therapies approved to date, the toxicity associated with them, and the advantages and limitations that they present as immunotherapy.
Collapse
Affiliation(s)
| | - Lilian Maritza Spencer
- School of Biological Sciences and Engineering, Yachay Tech University, San Miguel de Urcuquí, Ecuador
- Cell Biology Department, Simón Bolívar University, Valle de Sartenejas, Caracas, Venezuela
| |
Collapse
|
36
|
Cui Y, Luo M, Gu C, He Y, Yao Y, Li P. CAR designs for solid tumors: overcoming hurdles and paving the way for effective immunotherapy. BIOPHYSICS REPORTS 2023; 9:279-297. [PMID: 38516299 PMCID: PMC10951476 DOI: 10.52601/bpr.2023.230020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/28/2023] [Indexed: 03/23/2024] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy has revolutionized immunotherapy by modifying patients' immune cells genetically. By expressing CARs, these modified cells can specifically identify and eliminate tumor cells. The success of CAR-T therapy in hematological malignancies, such as leukemia and lymphoma, has been remarkable. Numerous studies have reported improved patient outcomes and increased survival rates. However, the application of CAR-T therapy in treating solid tumors faces significant challenges. Solid tumors possess complex microenvironments containing stromal cells, extracellular matrix components, and blood vessels. These factors can impede the infiltration and persistence of CAR-T cells within the tumor. Additionally, the lack of target antigens exclusively expressed on tumor cells raises concerns about off-target effects and potential toxicity. This review aims to discuss advancements achieved by CAR-T therapy in solid tumors and the clinical outcomes in the realm of solid tumors.
Collapse
Affiliation(s)
- Yuanbin Cui
- China-New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Mintao Luo
- China-New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Chuanyuan Gu
- China-New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yuxian He
- University of California San Diego, La Jolla, CA 92093-0021, USA
| | - Yao Yao
- China-New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Peng Li
- China-New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| |
Collapse
|
37
|
Wang M. Express Delivery of Next-Generation CAR T Cells with Preserved Naive and Stemness Phenotypes for the Treatment of Aggressive Lymphomas. Cancer Discov 2023; 13:1961-1963. [PMID: 37671474 DOI: 10.1158/2159-8290.cd-23-0735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
SUMMARY In this issue of Cancer Discovery, Dickinson and colleagues present clinical data from a first-in-human study of YTB323, a novel autologous CD19-directed chimeric antigen receptor T-cell therapy generated on the T-Charge platform with preserved naive state and stemness phenotypes. Treatment with YTB323 achieved high overall response rates, durable complete remissions, and good overall safety. Their cell doses are up to 25-fold lower than with tisagenlecleucel. See related article by Dickinson et al., p. 1982 (10).
Collapse
Affiliation(s)
- Michael Wang
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
38
|
Tapia-Galisteo A, Álvarez-Vallina L, Sanz L. Bi- and trispecific immune cell engagers for immunotherapy of hematological malignancies. J Hematol Oncol 2023; 16:83. [PMID: 37501154 PMCID: PMC10373336 DOI: 10.1186/s13045-023-01482-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023] Open
Abstract
Immune cell engagers are engineered antibodies with at least one arm binding a tumor-associated antigen and at least another one directed against an activating receptor in immune effector cells: CD3 for recruitment of T cells and CD16a for NK cells. The first T cell engager (the anti-CD19 blinatumomab) was approved by the FDA in 2014, but no other one hit the market until 2022. Now the field is gaining momentum, with three approvals in 2022 and 2023 (as of May): the anti-CD20 × anti-CD3 mosunetuzumab and epcoritamab and the anti-B cell maturation antigen (BCMA) × anti-CD3 teclistamab, and another three molecules in regulatory review. T cell engagers will likely revolutionize the treatment of hematological malignancies in the short term, as they are considerably more potent than conventional monoclonal antibodies recognizing the same tumor antigens. The field is thriving, with a plethora of different formats and targets, and around 100 bispecific T cell engagers more are already in clinical trials. Bispecific NK cell engagers are also in early-stage clinical studies and may offer similar efficacy with milder side effects. Trispecific antibodies (engaging either T cell or NK cell receptors) raise the game even further with a third binding moiety, which allows either the targeting of an additional tumor-associated antigen to increase specificity and avoid immune escape or the targeting of additional costimulatory receptors on the immune cell to improve its effector functions. Altogether, these engineered molecules may change the paradigm of treatment for relapsed or refractory hematological malignancies.
Collapse
Affiliation(s)
- Antonio Tapia-Galisteo
- Immuno-Oncology and Immunotherapy Group, Biomedical Research Institute Hospital Universitario, 12 de Octubre, Madrid, Spain
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario, 12 de Octubre, Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Luis Álvarez-Vallina
- Immuno-Oncology and Immunotherapy Group, Biomedical Research Institute Hospital Universitario, 12 de Octubre, Madrid, Spain.
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario, 12 de Octubre, Madrid, Spain.
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain.
| | - Laura Sanz
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain.
| |
Collapse
|
39
|
Westin JR, Oluwole OO, Kersten MJ, Miklos DB, Perales MA, Ghobadi A, Rapoport AP, Sureda A, Jacobson CA, Farooq U, van Meerten T, Ulrickson M, Elsawy M, Leslie LA, Chaganti S, Dickinson M, Dorritie K, Reagan PM, McGuirk J, Song KW, Riedell PA, Minnema MC, Yang Y, Vardhanabhuti S, Filosto S, Cheng P, Shahani SA, Schupp M, To C, Locke FL. Survival with Axicabtagene Ciloleucel in Large B-Cell Lymphoma. N Engl J Med 2023; 389:148-157. [PMID: 37272527 DOI: 10.1056/nejmoa2301665] [Citation(s) in RCA: 197] [Impact Index Per Article: 98.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
BACKGROUND In an analysis of the primary outcome of this phase 3 trial, patients with early relapsed or refractory large B-cell lymphoma who received axicabtagene ciloleucel (axi-cel), an autologous anti-CD19 chimeric antigen receptor T-cell therapy, as second-line treatment had significantly longer event-free survival than those who received standard care. Data were needed on longer-term outcomes. METHODS In this trial, we randomly assigned patients with early relapsed or refractory large B-cell lymphoma in a 1:1 ratio to receive either axi-cel or standard care (two to three cycles of chemoimmunotherapy followed by high-dose chemotherapy with autologous stem-cell transplantation in patients who had a response). The primary outcome was event-free survival, and key secondary outcomes were response and overall survival. Here, we report the results of the prespecified overall survival analysis at 5 years after the first patient underwent randomization. RESULTS A total of 359 patients underwent randomization to receive axi-cel (180 patients) or standard care (179 patients). At a median follow-up of 47.2 months, death had been reported in 82 patients in the axi-cel group and in 95 patients in the standard-care group. The median overall survival was not reached in the axi-cel group and was 31.1 months in the standard-care group; the estimated 4-year overall survival was 54.6% and 46.0%, respectively (hazard ratio for death, 0.73; 95% confidence interval [CI], 0.54 to 0.98; P = 0.03 by stratified two-sided log-rank test). This increased survival with axi-cel was observed in the intention-to-treat population, which included 74% of patients with primary refractory disease and other high-risk features. The median investigator-assessed progression-free survival was 14.7 months in the axi-cel group and 3.7 months in the standard-care group, with estimated 4-year percentages of 41.8% and 24.4%, respectively (hazard ratio, 0.51; 95% CI, 0.38 to 0.67). No new treatment-related deaths had occurred since the primary analysis of event-free survival. CONCLUSIONS At a median follow-up of 47.2 months, axi-cel as second-line treatment for patients with early relapsed or refractory large B-cell lymphoma resulted in significantly longer overall survival than standard care. (Funded by Kite; ZUMA-7 ClinicalTrials.gov number, NCT03391466.).
Collapse
Affiliation(s)
- Jason R Westin
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Olalekan O Oluwole
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Marie José Kersten
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - David B Miklos
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Miguel-Angel Perales
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Armin Ghobadi
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Aaron P Rapoport
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Anna Sureda
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Caron A Jacobson
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Umar Farooq
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Tom van Meerten
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Matthew Ulrickson
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Mahmoud Elsawy
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Lori A Leslie
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Sridhar Chaganti
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Michael Dickinson
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Kathleen Dorritie
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Patrick M Reagan
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Joseph McGuirk
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Kevin W Song
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Peter A Riedell
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Monique C Minnema
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Yin Yang
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Saran Vardhanabhuti
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Simone Filosto
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Paul Cheng
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Shilpa A Shahani
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Marco Schupp
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Christina To
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Frederick L Locke
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| |
Collapse
|
40
|
Ziegler N, Cortés-López M, Alt F, Sprang M, Ustjanzew A, Lehmann N, El Malki K, Wingerter A, Russo A, Beck O, Attig S, Roth L, König J, Paret C, Faber J. Analysis of RBP expression and binding sites identifies PTBP1 as a regulator of CD19 expression in B-ALL. Oncoimmunology 2023; 12:2184143. [PMID: 36875548 PMCID: PMC9980455 DOI: 10.1080/2162402x.2023.2184143] [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: 03/05/2023] Open
Abstract
Despite massive improvements in the treatment of B-ALL through CART-19 immunotherapy, a large number of patients suffer a relapse due to loss of the targeted epitope. Mutations in the CD19 locus and aberrant splicing events are known to account for the absence of surface antigen. However, early molecular determinants suggesting therapy resistance as well as the time point when first signs of epitope loss appear to be detectable are not enlightened so far. By deep sequencing of the CD19 locus, we identified a blast-specific 2-nucleotide deletion in intron 2 that exists in 35% of B-ALL samples at initial diagnosis. This deletion overlaps with the binding site of RNA binding proteins (RBPs) including PTBP1 and might thereby affect CD19 splicing. Moreover, we could identify a number of other RBPs that are predicted to bind to the CD19 locus being deregulated in leukemic blasts, including NONO. Their expression is highly heterogeneous across B-ALL molecular subtypes as shown by analyzing 706 B-ALL samples accessed via the St. Jude Cloud. Mechanistically, we show that downregulation of PTBP1, but not of NONO, in 697 cells reduces CD19 total protein by increasing intron 2 retention. Isoform analysis in patient samples revealed that blasts, at diagnosis, express increased amounts of CD19 intron 2 retention compared to normal B cells. Our data suggest that loss of RBP functionality by mutations altering their binding motifs or by deregulated expression might harbor the potential for the disease-associated accumulation of therapy-resistant CD19 isoforms.
Collapse
Affiliation(s)
- Nicole Ziegler
- Center for Pediatric and Adolescent Medicine, Department of Pediatric Hematology/Oncology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | | | - Francesca Alt
- Center for Pediatric and Adolescent Medicine, Department of Pediatric Hematology/Oncology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Maximilian Sprang
- Faculty of Biology, Johannes Gutenberg University Mainz, Biozentrum I, Mainz, Germany
| | - Arsenij Ustjanzew
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Nadine Lehmann
- Center for Pediatric and Adolescent Medicine, Department of Pediatric Hematology/Oncology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Khalifa El Malki
- Center for Pediatric and Adolescent Medicine, Department of Pediatric Hematology/Oncology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Arthur Wingerter
- Center for Pediatric and Adolescent Medicine, Department of Pediatric Hematology/Oncology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Alexandra Russo
- Center for Pediatric and Adolescent Medicine, Department of Pediatric Hematology/Oncology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Olaf Beck
- Center for Pediatric and Adolescent Medicine, Department of Pediatric Hematology/Oncology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sebastian Attig
- Department of Translational Oncology and Immunology at the Institute of Immunology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lea Roth
- Center for Pediatric and Adolescent Medicine, Department of Pediatric Hematology/Oncology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Julian König
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Claudia Paret
- Center for Pediatric and Adolescent Medicine, Department of Pediatric Hematology/Oncology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,German Cancer Consortium (DKTK), Site Frankfurt/Mainz, Germany, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jörg Faber
- Center for Pediatric and Adolescent Medicine, Department of Pediatric Hematology/Oncology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,German Cancer Consortium (DKTK), Site Frankfurt/Mainz, Germany, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|