1
|
Choi S, Hossain M, Lee H, Baek J, Park HS, Lim CL, Han D, Park T, Kim JH, Gong G, Kweon MN, Lee HJ. Expansion of tumor-infiltrating lymphocytes from head and neck squamous cell carcinoma to assess the potential of adoptive cell therapy. Cancer Immunol Immunother 2024; 73:101. [PMID: 38630265 PMCID: PMC11024072 DOI: 10.1007/s00262-024-03691-9] [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: 02/01/2024] [Accepted: 03/25/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Adoptive transfer of in vitro expanded tumor-infiltrating lymphocytes (TILs) has been effective in regressing several types of malignant tumors. This study assessed the yield and factors influencing the successful expansion of tumor-infiltrating lymphocytes (TILs) from head and neck squamous cell carcinoma (HNSCC), along with their immune phenotypes. METHODS TILs were expanded from 47 surgically resected HNSCC specimens and their metastasized lymph nodes. The cancer tissues were cut into small pieces (1-2 mm) and underwent initial expansion for 2 weeks. Tumor location, smoking history, stromal TIL percentage, human papillomavirus infection, and programmed death-ligand 1 score were examined for their impact on successful expansion of TILs. Expanded TILs were evaluated by flow cytometry using fluorescence-activated cell sorting. A second round of TIL expansion following the rapid expansion protocol was performed on a subset of samples with successful TIL expansion. RESULTS TILs were successfully expanded from 36.2% samples. Failure was due to contamination (27.6%) or insufficient expansion (36.2%). Only the stromal TIL percentage was significantly associated with successful TIL expansion (p = 0.032). The stromal TIL percentage also displayed a correlation with the expanded TILs per fragment (r = 0.341, p = 0.048). On flow cytometry analysis using 13 samples with successful TIL expansion, CD4 + T cell dominancy was seen in 69.2% of cases. Effector memory T cells were the major phenotype of expanded CD4 + and CD8 + T cells in all cases. CONCLUSION We could expand TILs from approximately one-third of HNSCC samples. TIL expansion could be applicable in HNSCC samples with diverse clinicopathological characteristics.
Collapse
Affiliation(s)
- Sangjoon Choi
- Department of Pathology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro, 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Mofazzal Hossain
- Department of Medical Science, Brain Korea 21 project, AMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hyun Lee
- Department of Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jina Baek
- Department of Pathology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro, 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | | | | | - DoYeon Han
- Department of Medical Science, Brain Korea 21 project, AMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Taehyun Park
- Department of Medical Science, Brain Korea 21 project, AMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jong Hyeok Kim
- Department of Medical Science, Brain Korea 21 project, AMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Gyungyub Gong
- Department of Pathology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro, 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| | - Mi-Na Kweon
- Mucosal Immunology Laboratory, Department of Convergence Medicine, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| | - Hee Jin Lee
- Department of Pathology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro, 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
- NeogenTC Corp, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Liu Y, Wang T, Ma W, Jia Z, Wang Q, Zhang M, Luo Y, Sun H. Metabolic reprogramming in the tumor microenvironment: unleashing T cell stemness for enhanced cancer immunotherapy. Front Pharmacol 2023; 14:1327717. [PMID: 38169800 PMCID: PMC10758489 DOI: 10.3389/fphar.2023.1327717] [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: 10/25/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024] Open
Abstract
T cells play a pivotal role in the immune system by distinguishing between various harmful pathogens and cancerous cells within the human body and initiating an immune response. Within the tumor microenvironment (TME), immune effector T cells encounter both immunosuppressive cells and factors that hinder their functionality. Additionally, they endure robust and persistent antigenic stimulation, often leading to exhaustion and apoptosis. However, the stemness of T cells, characterized by their ability to survive and self-renew over extended periods, represents a primary target in immune checkpoint therapies such as anti-PD-1 therapy. T cell stemness encompasses specific memory T cell subsets and progenitor-exhausted T cells with stem cell-like properties. Therefore, understanding the impact of the TME on T cell stemness, including factors like K+, lactate, and H+, holds significant importance and can facilitate the mitigation of terminal T-cell depletion, the identification of potential resilient biomarkers or therapeutic targets resistant to immune checkpoint therapies, and ultimately lead to sustained anti-tumor effects. Thus, it offers a novel perspective for advancing tumor immunotherapy.
Collapse
Affiliation(s)
- Youhan Liu
- College of Sport and Health, Shandong Sport University, Jinan, China
| | - Tao Wang
- Department of Pediatric Surgery, Zibo Central Hospital, Zibo, China
| | - Wen Ma
- College of Sport and Health, Shandong Sport University, Jinan, China
| | - Zixuan Jia
- College of Sport and Health, Shandong Sport University, Jinan, China
| | - Qinglu Wang
- College of Sport and Health, Shandong Sport University, Jinan, China
| | - Maoling Zhang
- College of Sport and Health, Shandong Sport University, Jinan, China
| | - Ying Luo
- Department of Clinical Laboratory, Zibo Central Hospital, Zibo, China
| | - Hongmei Sun
- College of Sport and Health, Shandong Sport University, Jinan, China
| |
Collapse
|
3
|
Marchais M, Simula L, Phayanouvong M, Mami-Chouaib F, Bismuth G, Decroocq J, Bouscary D, Dutrieux J, Mangeney M. FOXO1 Inhibition Generates Potent Nonactivated CAR T Cells against Solid Tumors. Cancer Immunol Res 2023; 11:1508-1523. [PMID: 37649096 DOI: 10.1158/2326-6066.cir-22-0533] [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: 07/04/2022] [Revised: 01/09/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023]
Abstract
Chimeric antigen receptor (CAR) T cells have shown promising results in the treatment of B-cell malignancies. Despite the successes, challenges remain. One of them directly involves the CAR T-cell manufacturing process and especially the ex vivo activation phase. While this is required to allow infection and expansion, ex vivo activation dampens the antitumor potential of CAR T cells. Optimizing the nature of the T cells harboring the CAR is a strategy to address this obstacle and has the potential to improve CAR T-cell therapy, including for solid tumors. Here, we describe a protocol to create CAR T cells without ex vivo preactivation by inhibiting the transcription factor FOXO1 (CAR TAS cells). This approach made T cells directly permissive to lentiviral infection, allowing CAR expression, with enhanced antitumor functions. FOXO1 inhibition in primary T cells (TAS cells) correlated with acquisition of a stem cell memory phenotype, high levels of granzyme B, and increased production of TNFα. TAS cells displayed enhanced proliferative and cytotoxic capacities as well as improved migratory properties. In vivo experiments showed that CAR TAS cells were more efficient at controlling solid tumor growth than classical CAR T cells. The production of CAR TAS from patients' cells confirmed the feasibility of the protocol in clinic.
Collapse
Affiliation(s)
- Maude Marchais
- CNRS UMR9196, Physiologie et Pathologie Moléculaires des Rétrovirus Endogènes et Infectieux, Gustave Roussy, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
- Université de Paris, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
| | - Luca Simula
- Université de Paris, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
| | - Mélanie Phayanouvong
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Fathia Mami-Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Georges Bismuth
- Université de Paris, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
| | - Justine Decroocq
- Assistance Publique-Hôpitaux de Paris, Centre-Université de Paris, Service d'Hématologie Clinique, Hôpital Cochin, Paris, France
| | - Didier Bouscary
- Université de Paris, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
- Assistance Publique-Hôpitaux de Paris, Centre-Université de Paris, Service d'Hématologie Clinique, Hôpital Cochin, Paris, France
| | - Jacques Dutrieux
- Université de Paris, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
- Viral DNA Integration and Chromatin Dynamics Network (DyNAVir), Paris, France
| | - Marianne Mangeney
- CNRS UMR9196, Physiologie et Pathologie Moléculaires des Rétrovirus Endogènes et Infectieux, Gustave Roussy, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
- Université de Paris, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
| |
Collapse
|
4
|
Feng W, He Z, Shi L, Zhu Z, Ma H. Significance of CD80 as a Prognostic and Immunotherapeutic Biomarker in Lung Adenocarcinoma. Biochem Genet 2023; 61:1937-1966. [PMID: 36892747 PMCID: PMC10517904 DOI: 10.1007/s10528-023-10343-7] [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: 11/30/2022] [Accepted: 02/02/2023] [Indexed: 03/10/2023]
Abstract
Lung adenocarcinoma (LUAD) is the primary cause of death among pulmonary cancer patients. Upregulation of CD80 may interact with cytotoxic T lymphocyte antigen 4 (CTLA4) to promote tumor progression and provide a potential target for biological antitumor therapy. However, the role of CD80 in LUAD is still unclear. To investigate the function of CD80 in LUAD, we collected transcriptomic data from 594 lung samples from The Cancer Genome Atlas of America (TCGA) database, along with the corresponding clinical information. We systematically explored the role of CD80 in LUAD using bioinformatics methods, including GO enrichment analysis, KEGG pathway analysis, Gene Set Enrichment Analysis (GSEA), co-expression analysis, and the CIBERSORT algorithm. Finally, we investigated the differences between the two subgroups of CD80 expression in terms of some drug sensitivity, using the pRRophetic package to screen small molecular drugs for therapeutic use. A predictive model based on CD80 for LUAD patients was successfully constructed. In addition, we discovered that the CD80-based prediction model was an independent prognostic factor. Co-expression analysis revealed 10 CD80-related genes, including oncogenes and immune-related genes. Functional analysis showed that the differentially expressed genes in patients with high CD80 expression were mainly located in immune-related signaling pathways. CD80 expression was also associated with immune cell infiltration and immune checkpoints. Highly expressing patients were more sensitive to several drugs, such as rapamycin, paclitaxel, crizotinib, and bortezomib. Finally, we found evidence that 15 different small molecular drugs may benefit the treatment of LUAD patients. This study found that elevated CD80 pairs could improve the prognosis of LUAD patients. CD80 is likely to be a potential as a prognostic and therapeutic target. The future use of small molecular drugs in combination with immune checkpoint blockade to enhance antitumor therapy and improve prognosis for LUAD patients is promising.
Collapse
Affiliation(s)
- Wei Feng
- First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ziyi He
- First Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Shi
- First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zheng Zhu
- First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haitao Ma
- First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
5
|
Mehra V, Agliardi G, Dias Alves Pinto J, Shafat MS, Garai AC, Green L, Hotblack A, Arce Vargas F, Peggs KS, van der Waart AB, Dolstra H, Pule MA, Roddie C. AKT inhibition generates potent polyfunctional clinical grade AUTO1 CAR T-cells, enhancing function and survival. J Immunother Cancer 2023; 11:e007002. [PMID: 37709295 PMCID: PMC10503365 DOI: 10.1136/jitc-2023-007002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND AUTO1 is a fast off-rate CD19-targeting chimeric antigen receptor (CAR), which has been successfully tested in adult lymphoblastic leukemia. Tscm/Tcm-enriched CAR-T populations confer the best expansion and persistence, but Tscm/Tcm numbers are poor in heavily pretreated adult patients. To improve this, we evaluate the use of AKT inhibitor (VIII) with the aim of uncoupling T-cell expansion from differentiation, to enrich Tscm/Tcm subsets. METHODS VIII was incorporated into the AUTO1 manufacturing process based on the semiautomated the CliniMACS Prodigy platform at both small and cGMP scale. RESULTS AUTO1 manufactured with VIII showed Tscm/Tcm enrichment, improved expansion and cytotoxicity in vitro and superior antitumor activity in vivo. Further, VIII induced AUTO1 Th1/Th17 skewing, increased polyfunctionality, and conferred a unique metabolic profile and a novel signature for autophagy to support enhanced expansion and cytotoxicity. We show that VIII-cultured AUTO1 products from B-ALL patients on the ALLCAR19 study possess superior phenotype, metabolism, and function than parallel control products and that VIII-based manufacture is scalable to cGMP. CONCLUSION Ultimately, AUTO1 generated with VIII may begin to overcome the product specific factors contributing to CD19+relapse.
Collapse
Affiliation(s)
- Vedika Mehra
- Research Department of Haematology, University College London, London, UK
| | - Giulia Agliardi
- Research Department of Haematology, University College London, London, UK
- Centre for Cell, Gene and Tissue Therapeutics, Royal Free Hospital, London, UK
| | - Juliana Dias Alves Pinto
- Research Department of Haematology, University College London, London, UK
- Centre for Cell, Gene and Tissue Therapeutics, Royal Free Hospital, London, UK
| | - Manar S Shafat
- Research Department of Haematology, University College London, London, UK
| | | | - Louisa Green
- Research Department of Haematology, University College London, London, UK
| | - Alastair Hotblack
- Research Department of Haematology, University College London, London, UK
| | | | - Karl S Peggs
- Research Department of Haematology, University College London, London, UK
| | - Anniek B van der Waart
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Harry Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Martin A Pule
- Research Department of Haematology, University College London, London, UK
- Autolus Ltd, London, UK
| | - Claire Roddie
- Research Department of Haematology, University College London, London, UK
| |
Collapse
|
6
|
Chen R, Wu J, Liu S, Sun Y, Liu G, Zhang L, Yu Q, Xu J, Meng L. Immune-related risk prognostic model for clear cell renal cell carcinoma: Implications for immunotherapy. Medicine (Baltimore) 2023; 102:e34786. [PMID: 37653791 PMCID: PMC10470711 DOI: 10.1097/md.0000000000034786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 09/02/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is associated with complex immune interactions. We conducted a comprehensive analysis of immune-related differentially expressed genes in patients with ccRCC using data from The Cancer Genome Atlas and ImmPort databases. The immune-related differentially expressed genes underwent functional and pathway enrichment analysis, followed by COX regression combined with LASSO regression to construct an immune-related risk prognostic model. The model comprised 4 IRGs: CLDN4, SEMA3G, CAT, and UCN. Patients were stratified into high-risk and low-risk groups based on the median risk score, and the overall survival rate of the high-risk group was significantly lower than that of the low-risk group, confirming the reliability of the model from various perspectives. Further comparison of immune infiltration, tumor mutation load, and immunophenoscore (IPS) comparison between the 2 groups indicates that the high-risk group could potentially demonstrate a heightened sensitivity towards immunotherapy checkpoints PD-1, CTLA-4, IL-6, and LAG3 in ccRCC patients. The proposed model not only applies to ccRCC but also shows potential in developing into a prognostic model for renal cancer, thus introducing a novel approach for personalized immunotherapy in ccRCC.
Collapse
Affiliation(s)
- Ronghui Chen
- Clinical Medical College of Weifang Medical University, Weifang, China
| | - Jun Wu
- Department of Oncology, People’s Hospital of Rizhao, Rizhao, China
| | - Shan Liu
- Department of Oncology, People’s Hospital of Rizhao, Rizhao, China
| | - Yefeng Sun
- Department of Emergency, People’s Hospital of Rizhao, Rizhao, China
| | - Guozhi Liu
- Jining Medical University, Jining, China
| | - Lin Zhang
- Jining Medical University, Jining, China
| | - Qing Yu
- Clinical Medical College of Weifang Medical University, Weifang, China
| | - Juan Xu
- Clinical Medical College of Weifang Medical University, Weifang, China
| | - Lingxin Meng
- Department of Oncology, People’s Hospital of Rizhao, Rizhao, China
| |
Collapse
|
7
|
Cappell KM, Kochenderfer JN. Long-term outcomes following CAR T cell therapy: what we know so far. Nat Rev Clin Oncol 2023; 20:359-371. [PMID: 37055515 PMCID: PMC10100620 DOI: 10.1038/s41571-023-00754-1] [Citation(s) in RCA: 300] [Impact Index Per Article: 300.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2023] [Indexed: 04/15/2023]
Abstract
Chimeric antigen receptors (CAR) are engineered fusion proteins designed to target T cells to antigens expressed on cancer cells. CAR T cells are now an established treatment for patients with relapsed and/or refractory B cell lymphomas, B cell acute lymphoblastic leukaemia and multiple myeloma. At the time of this writing, over a decade of follow-up data are available from the initial patients who received CD19-targeted CAR T cells for B cell malignancies. Data on the outcomes of patients who received B cell maturation antigen (BCMA)-targeted CAR T cells for multiple myeloma are more limited owing to the more recent development of these constructs. In this Review, we summarize long-term follow-up data on efficacy and toxicities from patients treated with CAR T cells targeting CD19 or BCMA. Overall, the data demonstrate that CD19-targeted CAR T cells can induce prolonged remissions in patients with B cell malignancies, often with minimal long-term toxicities, and are probably curative for a subset of patients. By contrast, remissions induced by BCMA-targeted CAR T cells are typically more short-lived but also generally have only limited long-term toxicities. We discuss factors associated with long-term remissions, including the depth of initial response, malignancy characteristics predictive of response, peak circulating CAR levels and the role of lymphodepleting chemotherapy. We also discuss ongoing investigational strategies designed to improve the length of remission following CAR T cell therapy.
Collapse
Affiliation(s)
- Kathryn M Cappell
- Surgery Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, USA
| | - James N Kochenderfer
- Surgery Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, USA.
| |
Collapse
|
8
|
Heuser C, Renner K, Kreutz M, Gattinoni L. Targeting lactate metabolism for cancer immunotherapy - a matter of precision. Semin Cancer Biol 2023; 88:32-45. [PMID: 36496155 DOI: 10.1016/j.semcancer.2022.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022]
Abstract
Immune checkpoint inhibitors and adoptive T cell therapies have been valuable additions to the toolbox in the fight against cancer. These treatments have profoundly increased the number of patients with a realistic perspective toward a return to a cancer-free life. Yet, in a number of patients and tumor entities, cancer immunotherapies have been ineffective so far. In solid tumors, immune exclusion and the immunosuppressive tumor microenvironment represent substantial roadblocks to successful therapeutic outcomes. A major contributing factor to the depressed anti-tumor activity of immune cells in tumors is the harsh metabolic environment. Hypoxia, nutrient competition with tumor and stromal cells, and accumulating noxious waste products, including lactic acid, pose massive constraints to anti-tumor immune cells. Numerous strategies are being developed to exploit the metabolic vulnerabilities of tumor cells in the hope that these would also alleviate metabolism-inflicted immune suppression. While promising in principle, especially in combination with immunotherapies, these strategies need to be scrutinized for their effect on tumor-fighting immune cells, which share some of their key metabolic properties with tumor cells. Here, we provide an overview of strategies that seek to tackle lactate metabolism in tumor or immune cells to unleash anti-tumor immune responses, thereby opening therapeutic options for patients whose tumors are currently not treatable.
Collapse
Affiliation(s)
- Christoph Heuser
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy (LIT), 93053 Regensburg, Germany.
| | - Kathrin Renner
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany; Department of Otorhinolaryngology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Marina Kreutz
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany; Clinical Cooperation Group Immunometabolomics, Leibniz Institute for Immunotherapy (LIT), 93053 Regensburg, Germany; Center for Immunomedicine in Transplantation and Oncology (CITO), University Hospital Regensburg, 93053 Regensburg, Germany
| | - Luca Gattinoni
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy (LIT), 93053 Regensburg, Germany; Center for Immunomedicine in Transplantation and Oncology (CITO), University Hospital Regensburg, 93053 Regensburg, Germany; University of Regensburg, 93053 Regensburg, Germany.
| |
Collapse
|
9
|
Wilkens AB, Fulton EC, Pont MJ, Cole GO, Leung I, Stull SM, Hart MR, Bernstein ID, Furlan SN, Riddell SR. NOTCH1 signaling during CD4+ T-cell activation alters transcription factor networks and enhances antigen responsiveness. Blood 2022; 140:2261-2275. [PMID: 35605191 PMCID: PMC9837446 DOI: 10.1182/blood.2021015144] [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: 12/12/2021] [Accepted: 05/09/2022] [Indexed: 01/21/2023] Open
Abstract
Adoptive transfer of T cells expressing chimeric antigen receptors (CAR-T) effectively treats refractory hematologic malignancies in a subset of patients but can be limited by poor T-cell expansion and persistence in vivo. Less differentiated T-cell states correlate with the capacity of CAR-T to proliferate and mediate antitumor responses, and interventions that limit tumor-specific T-cell differentiation during ex vivo manufacturing enhance efficacy. NOTCH signaling is involved in fate decisions across diverse cell lineages and in memory CD8+ T cells was reported to upregulate the transcription factor FOXM1, attenuate differentiation, and enhance proliferation and antitumor efficacy in vivo. Here, we used a cell-free culture system to provide an agonistic NOTCH1 signal during naïve CD4+ T-cell activation and CAR-T production and studied the effects on differentiation, transcription factor expression, cytokine production, and responses to tumor. NOTCH1 agonism efficiently induced a stem cell memory phenotype in CAR-T derived from naïve but not memory CD4+ T cells and upregulated expression of AhR and c-MAF, driving heightened production of interleukin-22, interleukin-10, and granzyme B. NOTCH1-agonized CD4+ CAR-T demonstrated enhanced antigen responsiveness and proliferated to strikingly higher frequencies in mice bearing human lymphoma xenografts. NOTCH1-agonized CD4+ CAR-T also provided superior help to cotransferred CD8+ CAR-T, driving improved expansion and curative antitumor responses in vivo at low CAR-T doses. Our data expand the mechanisms by which NOTCH can shape CD4+ T-cell behavior and demonstrate that activating NOTCH1 signaling during genetic modification ex vivo is a potential strategy for enhancing the function of T cells engineered with tumor-targeting receptors.
Collapse
Affiliation(s)
- Alec B. Wilkens
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Molecular and Cellular Biology, University of Washington, Seattle, WA
| | - Elena C. Fulton
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Margot J. Pont
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Gabriel O. Cole
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Isabel Leung
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Sylvia M. Stull
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Matthew R. Hart
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Irwin D. Bernstein
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Scott N. Furlan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Stanley R. Riddell
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Molecular and Cellular Biology, University of Washington, Seattle, WA
| |
Collapse
|
10
|
Rial Saborido J, Völkl S, Aigner M, Mackensen A, Mougiakakos D. Role of CAR T Cell Metabolism for Therapeutic Efficacy. Cancers (Basel) 2022; 14:5442. [PMID: 36358860 PMCID: PMC9658570 DOI: 10.3390/cancers14215442] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/30/2022] [Accepted: 11/02/2022] [Indexed: 08/08/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cells hold enormous potential. However, a substantial proportion of patients receiving CAR T cells will not reach long-term full remission. One of the causes lies in their premature exhaustion, which also includes a metabolic anergy of adoptively transferred CAR T cells. T cell phenotypes that have been shown to be particularly well suited for CAR T cell therapy display certain metabolic characteristics; whereas T-stem cell memory (TSCM) cells, characterized by self-renewal and persistence, preferentially meet their energetic demands through oxidative phosphorylation (OXPHOS), effector T cells (TEFF) rely on glycolysis to support their cytotoxic function. Various parameters of CAR T cell design and manufacture co-determine the metabolic profile of the final cell product. A co-stimulatory 4-1BB domain promotes OXPHOS and formation of central memory T cells (TCM), while T cells expressing CARs with CD28 domains predominantly utilize aerobic glycolysis and differentiate into effector memory T cells (TEM). Therefore, modification of CAR co-stimulation represents one of the many strategies currently being investigated for improving CAR T cells' metabolic fitness and survivability within a hostile tumor microenvironment (TME). In this review, we will focus on the role of CAR T cell metabolism in therapeutic efficacy together with potential targets of intervention.
Collapse
Affiliation(s)
- Judit Rial Saborido
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Simon Völkl
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Michael Aigner
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität and University Hospital Erlangen, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Dimitrios Mougiakakos
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität and University Hospital Erlangen, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität and University Hospital Erlangen, 91054 Erlangen, Germany
- Medical Center, Department of Hematology and Oncology, Otto-von-Guericke University, 39120 Magdeburg, Germany
| |
Collapse
|
11
|
Fazeli P, Talepoor AG, Faghih Z, Gholijani N, Ataollahi MR, Ali‐Hassanzadeh M, Moravej H, Kalantar K. The frequency of CD4+ and CD8+ circulating T stem cell memory in type 1 diabetes. Immun Inflamm Dis 2022; 10:e715. [PMID: 36169248 PMCID: PMC9500591 DOI: 10.1002/iid3.715] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/27/2022] [Accepted: 09/13/2022] [Indexed: 11/07/2022] Open
Abstract
INTRODUCTION The frequencies and functions of T stem cell memory (TSCM) subsets vary in autoimmune diseases. We evaluated the frequencies of CD4+ and CD8+ TSCM subsets as well as their PD-1 expression levels in patients with T1D. METHODS Blood samples were collected from new case (NC) (n = 15), and long-term (LT) (n = 15) groups and healthy controls (n = 15). Five subsets of T cells including TCM(CD4+ /CD8+ CCR7+ CD45RO+ CD95+ ), TCMhi (CD4+ /CD8+ CCR7+ CD45ROhi CD95+ ), TEM(CD4+ /CD8+ CCR7- CD45RO+ CD95+ ), TSCM(CD4+ /CD8+ CCR7+ CD45RO- CD95+ ), and T naive (CD4+ /CD8+ CCR7+ CD45RO- CD95- ) were detected by flow-cytometry. RESULTS The frequency of CD4+ TSCM was higher in NC patients than LT patients and controls (p < .0001 and p = .0086, respectively). A higher percentage of the CD8+ T naive cells was shown in NC patients as compared with LT and healthy individuals (p = .0003 and p = .0002, respectively). An increased level of PD-1 expression was observed on the CD4+ TCM and TCMhi cells in LT patients as compared with healthy controls (p = .0037 and p = .0145, respectively). Also, the higher PD-1 expression was observed on the CD8+ TCM and TCMhi in NC and LT patients as compared with controls (p = .0068 and p < .0001; p = .0012 and p = .0012, respectively). CONCLUSION Considering TSCMs' capacities to generate all memory and effector T cells, our results may suggest a potential association between the increased frequencies of TSCMs and T1D progression.
Collapse
Affiliation(s)
- Pooriya Fazeli
- Department of ImmunologySchool of MedicineShiraz University of Medical SciencesShirazIran
| | - Atefe Ghamar Talepoor
- Department of ImmunologySchool of MedicineShiraz University of Medical SciencesShirazIran
| | - Zahra Faghih
- Shiraz Institute for Cancer ResearchSchool of MedicineShiraz University of Medical SciencesShirazIran
| | - Nasser Gholijani
- Autoimmune Diseases Research CenterShiraz University of Medical SciencesShirazIran
| | | | | | - Hossein Moravej
- Department of PediatricsSchool of MedicineShiraz University of Medical SciencesShirazIran
| | - Kurosh Kalantar
- Department of ImmunologySchool of MedicineShiraz University of Medical SciencesShirazIran
- Autoimmune Diseases Research CenterShiraz University of Medical SciencesShirazIran
| |
Collapse
|
12
|
Novel CD19 chimeric antigen receptor T cells manufactured next-day for acute lymphoblastic leukemia. Blood Cancer J 2022; 12:96. [PMID: 35750687 PMCID: PMC9232607 DOI: 10.1038/s41408-022-00688-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/23/2022] [Accepted: 05/31/2022] [Indexed: 11/23/2022] Open
Abstract
Chimeric antigen receptor-engineered T (CAR-T) cells have shown promising efficacy in patients with relapsed/refractory B cell acute lymphoblastic leukemia (R/R B-ALL). However, challenges remain including long manufacturing processes that need to be overcome. We presented the CD19-targeting CAR-T cell product GC007F manufactured next-day (FasTCAR-T cells) and administered to patients with R/R B-ALL. A total of 21 patients over 14 years of age with CD19+ R/R B-ALL were screened, enrolled and infused with a single infusion of GC007F CAR-T at three different dose levels. The primary objective of the study was to assess safety, secondary objectives included pharmacokinetics of GC007F cells in patients with R/R B-ALL and preliminary efficacy. We were able to demonstrate in preclinical studies that GC007F cells exhibited better proliferation and tumor killing than conventional CAR-T (C-CAR-T) cells. In this investigator-initiated study all 18 efficacy-evaluable patients achieved a complete remission (CR) (18/18, 100.00%) by day 28, with 17 of the patients (94.4%) achieving CR with minimal residual disease (MRD) negative. Fifteen (83.3%) remained disease free at the 3-month assessment, 14 patients (77.8%) maintaining MRD negative at month 3. Among all 21 enrolled patients, the median peak of CAR-T cell was on day 10, with a median peak copy number of 104899.5/µg DNA and a median persistence period of 56 days (range: 7–327 days). The incidence of cytokine release syndrome (CRS) was 95.2% (n = 20), with severe CRS occurring in 52.4% (n = 11) of the patients. Six patients (28.6%) developed neurotoxicity of any grade. GC007F demonstrated superior expansion capacity and a less exhausted phenotype as compared to (C-CAR-T) cells. Moreover, this first-in-human clinical study showed that the novel, next-day manufacturing FasTCAR-T cells was feasible with a manageable toxicity profile in patients with R/R B-ALL.
Collapse
|
13
|
'Stem-like' precursors are the fount to sustain persistent CD8 + T cell responses. Nat Immunol 2022; 23:836-847. [PMID: 35624209 DOI: 10.1038/s41590-022-01219-w] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 04/07/2022] [Indexed: 01/22/2023]
Abstract
Virus-specific CD8+ T cells that differentiate in the context of resolved versus persisting infections exhibit divergent phenotypic and functional characteristics, which suggests that their differentiation trajectories are governed by distinct cellular dynamics, developmental pathways and molecular mechanisms. For acute infection, it is long known that antigen-specific T cell populations contain terminally differentiated effector T cells, known as short-lived effector T cells, and proliferation-competent and differentiation-competent memory precursor T cells. More recently, it was identified that a similar functional segregation occurs in chronic infections. A failure to generate proliferation-competent precursor cells in chronic infections and tumors results in the collapse of the T cell response. Thus, these precursor cells are major therapeutic and prophylactic targets of immune interventions. These observations suggest substantial commonality between T cell responses in acute and chronic infections but there are also critical differences. We are therefore reviewing the common features and peculiarities of precursor cells in acute infections, different types of persistent infection and cancer.
Collapse
|
14
|
Alzawi A, Iftikhar A, Shalgm B, Jones S, Ellis I, Islam M. Receptor, Signal, Nucleus, Action: Signals That Pass through Akt on the Road to Head and Neck Cancer Cell Migration. Cancers (Basel) 2022; 14:2606. [PMID: 35681586 PMCID: PMC9179418 DOI: 10.3390/cancers14112606] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 02/06/2023] Open
Abstract
This review aims to provide evidence for the role of the tumour microenvironment in cancer progression, including invasion and metastasis. The tumour microenvironment is complex and consists of tumour cells and stromal-derived cells, in addition to a modified extracellular matrix. The cellular components synthesise growth factors such as EGF, TGFα and β, VEGF, and NGF, which have been shown to initiate paracrine signalling in head and neck cancer cells by binding to cell surface receptors. One example is the phosphorylation, and hence activation, of the signalling protein Akt, which can ultimately induce oral cancer cell migration in vitro. Blocking of Akt activation by an inhibitor, MK2206, leads to a significant decrease, in vitro, of cancer-derived cell migration, visualised in both wound healing and scatter assays. Signalling pathways have therefore been popular targets for the design of chemotherapeutic agents, but drug resistance has been observed and is related to direct tumour-tumour cell communication, the tumour-extracellular matrix interface, and tumour-stromal cell interactions. Translation of this knowledge to patient care is reliant upon a comprehensive understanding of the complex relationships present in the tumour microenvironment and could ultimately lead to the design of efficacious treatment regimens such as targeted therapy or novel therapeutic combinations.
Collapse
Affiliation(s)
| | | | | | | | | | - Mohammad Islam
- Unit of Cell & Molecular Biology, School of Dentistry, University of Dundee, Dundee DD1 4HN, UK; (A.A.); (A.I.); (B.S.); (S.J.); (I.E.)
| |
Collapse
|
15
|
Chakraborty P, Parikh RY, Choi S, Tran D, Gooz M, Hedley ZT, Kim DS, Pytel D, Kang I, Nadig SN, Beeson GC, Ball L, Mehrotra M, Wang H, Berto S, Palanisamy V, Li H, Chatterjee S, Rodriguez PC, Maldonado EN, Diehl JA, Gangaraju VK, Mehrotra S. Carbon Monoxide Activates PERK-Regulated Autophagy to Induce Immunometabolic Reprogramming and Boost Antitumor T-cell Function. Cancer Res 2022; 82:1969-1990. [PMID: 35404405 PMCID: PMC9117468 DOI: 10.1158/0008-5472.can-21-3155] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 01/27/2022] [Accepted: 03/17/2022] [Indexed: 11/16/2022]
Abstract
Mitochondria and endoplasmic reticulum (ER) share structural and functional networks and activate well-orchestrated signaling processes to shape cells' fate and function. While persistent ER stress (ERS) response leads to mitochondrial collapse, moderate ERS promotes mitochondrial function. Strategies to boost antitumor T-cell function by targeting ER-mitochondria cross-talk have not yet been exploited. Here, we used carbon monoxide (CO), a short-lived gaseous molecule, to test whether engaging moderate ERS conditions can improve mitochondrial and antitumor functions in T cells. In melanoma antigen-specific T cells, CO-induced transient activation of ERS sensor protein kinase R-like endoplasmic reticulum kinase (PERK) significantly increased antitumor T-cell function. Furthermore, CO-induced PERK activation temporarily halted protein translation and induced protective autophagy, including mitophagy. The use of LC3-GFP enabled differentiation between the cells that prepare themselves to undergo active autophagy (LC3-GFPpos) and those that fail to enter the process (LC3-GFPneg). LC3-GFPpos T cells showed strong antitumor potential, whereas LC3-GFPneg cells exhibited a T regulatory-like phenotype, harbored dysfunctional mitochondria, and accumulated abnormal metabolite content. These anomalous ratios of metabolites rendered the cells with a hypermethylated state and distinct epigenetic profile, limiting their antitumor activity. Overall, this study shows that ERS-activated autophagy pathways modify the mitochondrial function and epigenetically reprogram T cells toward a superior antitumor phenotype to achieve robust tumor control. SIGNIFICANCE Transient activation of ER stress with carbon monoxide drives mitochondrial biogenesis and protective autophagy that elicits superior antitumor T-cell function, revealing an approach to improving adoptive cell efficacy therapy.
Collapse
Affiliation(s)
- Paramita Chakraborty
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Rasesh Y Parikh
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Seungho Choi
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Danh Tran
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Monika Gooz
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Zachariah T Hedley
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Do-Sung Kim
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Dariusz Pytel
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Lodz, Poland
| | - Inhong Kang
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Satish N Nadig
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Gyda C Beeson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Lauren Ball
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina
| | - Meenal Mehrotra
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Stefano Berto
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Viswanathan Palanisamy
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Hong Li
- Department of Public Health, Medical University of South Carolina, Charleston, South Carolina
| | - Shilpak Chatterjee
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Paulo C Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Eduardo N Maldonado
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - J Alan Diehl
- Department of Biochemistry, Case Western University, Cleveland, Ohio
| | - Vamsi K Gangaraju
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
- Department of Microbiology & Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
16
|
Anti-Gr-1 Antibody Provides Short-Term Depletion of MDSC in Lymphodepleted Mice with Active-Specific Melanoma Therapy. Vaccines (Basel) 2022; 10:vaccines10040560. [PMID: 35455309 PMCID: PMC9032646 DOI: 10.3390/vaccines10040560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 03/24/2022] [Accepted: 03/26/2022] [Indexed: 11/28/2022] Open
Abstract
Lymphodepletion, reconstitution and active-specific tumor cell vaccination (LRAST) enhances the induction of tumor-specific T cells in a murine melanoma model. Myeloid-derived suppressor cells (MDSC) may counteract the induction of tumor-reactive T cells and their therapeutic efficacy. Thus, the aim of the study was to evaluate a possible benefit of MDSC depletion using anti-Gr-1 antibodies (Ab) in combination with LRAST. Female C57BL/6 mice with 3 days established subcutaneous (s.c.) D5 melanoma were lymphodepleted with cyclophosphamide and reconstituted with naive splenocytes. Vaccination was performed with irradiated syngeneic mGM-CSF-secreting D5G6 melanoma cells. MDSC depletion was performed using anti-Gr-1 Ab (clone RB6-8C5). Induction of tumor-specific T cells derived from tumor vaccine draining lymph nodes (TVDLN) was evaluated by the amount of tumor-specific interferon (IFN)-γ release. LRAST combined with anti-Gr-1 mAb administration enhanced the induction of tumor-specific T cells in TVDLN capable of releasing IFN-γ in a tumor-specific manner. Additional anti-Gr-1 mAb administration in LRAST-treated mice delayed growth of D5 melanomas by two weeks. Furthermore, we elucidate the impact of anti-Gr-1-depleting antibodies on the memory T cell compartment. Our data indicate that standard of care treatment regimens against cancer can be improved by implementing agents, e.g., depleting antibodies, which target and eliminate MDSC.
Collapse
|
17
|
Targeting oncogene and non-oncogene addiction to inflame the tumour microenvironment. Nat Rev Drug Discov 2022; 21:440-462. [PMID: 35292771 DOI: 10.1038/s41573-022-00415-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2022] [Indexed: 12/12/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the clinical management of multiple tumours. However, only a few patients respond to ICIs, which has generated considerable interest in the identification of resistance mechanisms. One such mechanism reflects the ability of various oncogenic pathways, as well as stress response pathways required for the survival of transformed cells (a situation commonly referred to as 'non-oncogene addiction'), to support tumour progression not only by providing malignant cells with survival and/or proliferation advantages, but also by establishing immunologically 'cold' tumour microenvironments (TMEs). Thus, both oncogene and non-oncogene addiction stand out as promising targets to robustly inflame the TME and potentially enable superior responses to ICIs.
Collapse
|
18
|
Jalili A, Hajifathali A, Bereimipour A, Roshandel E, Aghdami N. The Impact of Different Cell Culture Mediums on CD8+ T Cells Expansion: A Bioinformatics Study. CELL JOURNAL 2022; 24:155-162. [PMID: 35451586 PMCID: PMC9035229 DOI: 10.22074/cellj.2022.7779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 02/15/2021] [Indexed: 11/04/2022]
Abstract
Objective Different Cell Culture medias can affect the expansion of T cells. The aim of this study is to assess signaling pathways, protein interactions and genes in T cells cultured in different common T cell expansion medias to select the best candidate. Materials and Methods In this in silico observational study, with the use of bioinformatics analysis and the use of enrichment databases, gene expression profiles were investigated using microarray analysis. Results The results of this study were the joint selection of 26 upregulated genes and 59 downregulated genes that were involved in SREBP control of lipid synthesis, co-stimulatory signal during T-cell activation mitosis and chromosome dynamics, telomeres, telomerase, and cellular aging signal pathways. Conclusion Using bioinformatics analyzes, integrated and regular genes were selected as common genes CD80, LST1, ATM and ITM2B 4-1BBL, Akt inhibitor, interleukin 7 and 15 expansion media.
Collapse
Affiliation(s)
- Arsalan Jalili
- Department of Applied Cell Sciences, Faculty of Basic Sciences and Advanced Medical Technologies, Royan Institute, ACECR, Tehran,
Iran,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran
| | - Abbas Hajifathali
- Hematopoeitic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Bereimipour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran ,Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Elham Roshandel
- Hematopoeitic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran ,P.O.Box: 1985711151Hematopoeitic Stem Cell Research CenterShahid Beheshti University of Medical SciencesTehranIranP.O.Box: 16635-148Department of Regenerative MedicineCell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyACECRTehranIran
Emails:,
| | - Nasser Aghdami
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR,
Tehran, Iran ,P.O.Box: 1985711151Hematopoeitic Stem Cell Research CenterShahid Beheshti University of Medical SciencesTehranIranP.O.Box: 16635-148Department of Regenerative MedicineCell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyACECRTehranIran
Emails:,
| |
Collapse
|
19
|
Ghassemi S, Durgin JS, Nunez-Cruz S, Patel J, Leferovich J, Pinzone M, Shen F, Cummins KD, Plesa G, Cantu VA, Reddy S, Bushman FD, Gill SI, O'Doherty U, O'Connor RS, Milone MC. Rapid manufacturing of non-activated potent CAR T cells. Nat Biomed Eng 2022; 6:118-128. [PMID: 35190680 PMCID: PMC8860360 DOI: 10.1038/s41551-021-00842-6] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 12/17/2021] [Indexed: 12/14/2022]
Abstract
Chimaeric antigen receptor (CAR) T cells can generate durable clinical responses in B-cell haematologic malignancies. The manufacturing of these T cells typically involves their activation, followed by viral transduction and expansion ex vivo for at least 6 days. However, the activation and expansion of CAR T cells leads to their progressive differentiation and the associated loss of anti-leukaemic activity. Here we show that functional CAR T cells can be generated within 24 hours from T cells derived from peripheral blood without the need for T-cell activation or ex vivo expansion, and that the efficiency of viral transduction in this process is substantially influenced by the formulation of the medium and the surface area-to-volume ratio of the culture vessel. In mouse xenograft models of human leukaemias, the rapidly generated non-activated CAR T cells exhibited higher anti-leukaemic in vivo activity per cell than the corresponding activated CAR T cells produced using the standard protocol. The rapid manufacturing of CAR T cells may reduce production costs and broaden their applicability.
Collapse
Affiliation(s)
- Saba Ghassemi
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Joseph S Durgin
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Selene Nunez-Cruz
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jai Patel
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Leferovich
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marilia Pinzone
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Feng Shen
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katherine D Cummins
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gabriela Plesa
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vito Adrian Cantu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shantan Reddy
- 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
| | - Saar I Gill
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Una O'Doherty
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Roddy S O'Connor
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael C Milone
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
20
|
Ahmad Z, Somanath PR. AKT Isoforms in the Immune Response in Cancer. Curr Top Microbiol Immunol 2022; 436:349-366. [PMID: 36243852 DOI: 10.1007/978-3-031-06566-8_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
AKT is a protein kinase that exists in three isoforms: AKT1, AKT2, and AKT3. Though similar in structure, these isoforms display different effects. AKT is activated downstream of PI3K, and together, this signaling pathway helps regulate cellular processes including cell growth, proliferation, metabolism, survival, and apoptosis. Disruption in these pathways has been associated with disorders including cardiovascular diseases, developmental disorders, inflammatory responses, autoimmune diseases, neurologic disorders, type 2 diabetes, and several cancers. In cancer, deregulation in the PI3K/AKT pathway can be manifested as tumorigenesis, pathological angiogenesis, and metastasis. Increased activity has been correlated with tumor progression and resistance to cancer treatments. Recent studies have suggested that inhibition of the PI3K/AKT pathway plays a significant role in the development, expansion, and proliferation of cells of the immune system. Additionally, AKT has been found to play an important role in differentiating regulatory T cells, activating B cells, and augmenting tumor immunosurveillance. This emphasizes AKT as a potential target for inhibition in cancer therapy. This chapter reviews AKT structure and regulation, its different isoforms, its role in immune cells, and its modulation in oncotherapy.
Collapse
Affiliation(s)
- Zayd Ahmad
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA.
- Georgia Cancer Center, Vascular Biology Center and Department of Medicine, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
21
|
He Y, Sun MM, Zhang GG, Yang J, Chen KS, Xu WW, Li B. Targeting PI3K/Akt signal transduction for cancer therapy. Signal Transduct Target Ther 2021; 6:425. [PMID: 34916492 PMCID: PMC8677728 DOI: 10.1038/s41392-021-00828-5] [Citation(s) in RCA: 491] [Impact Index Per Article: 163.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 11/02/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt pathway plays a crucial role in various cellular processes and is aberrantly activated in cancers, contributing to the occurrence and progression of tumors. Examining the upstream and downstream nodes of this pathway could allow full elucidation of its function. Based on accumulating evidence, strategies targeting major components of the pathway might provide new insights for cancer drug discovery. Researchers have explored the use of some inhibitors targeting this pathway to block survival pathways. However, because oncogenic PI3K pathway activation occurs through various mechanisms, the clinical efficacies of these inhibitors are limited. Moreover, pathway activation is accompanied by the development of therapeutic resistance. Therefore, strategies involving pathway inhibitors and other cancer treatments in combination might solve the therapeutic dilemma. In this review, we discuss the roles of the PI3K/Akt pathway in various cancer phenotypes, review the current statuses of different PI3K/Akt inhibitors, and introduce combination therapies consisting of signaling inhibitors and conventional cancer therapies. The information presented herein suggests that cascading inhibitors of the PI3K/Akt signaling pathway, either alone or in combination with other therapies, are the most effective treatment strategy for cancer.
Collapse
Affiliation(s)
- Yan He
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Miao Miao Sun
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Tumor Pathology, Zhengzhou, China
| | - Guo Geng Zhang
- MOE Key Laboratory of Tumor Molecular Biology and Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jing Yang
- MOE Key Laboratory of Tumor Molecular Biology and Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Kui Sheng Chen
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Tumor Pathology, Zhengzhou, China.
| | - Wen Wen Xu
- MOE Key Laboratory of Tumor Molecular Biology and Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.
| | - Bin Li
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China.
| |
Collapse
|
22
|
Zhang M, Jin X, Sun R, Xiong X, Wang J, Xie D, Zhao M. Optimization of metabolism to improve efficacy during CAR-T cell manufacturing. J Transl Med 2021; 19:499. [PMID: 34876185 PMCID: PMC8650271 DOI: 10.1186/s12967-021-03165-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
Chimeric antigen receptor T cell (CAR-T cell) therapy is a relatively new, effective, and rapidly evolving therapeutic for adoptive immunotherapies. Although it has achieved remarkable effect in hematological malignancies, there are some problems that remain to be resolved. For example, there are high recurrence rates and poor efficacy in solid tumors. In this review, we first briefly describe the metabolic re-editing of T cells and the changes in metabolism during the preparation of CAR-T cells. Furthermore, we summarize the latest developments and newest strategies to improve the metabolic adaptability and antitumor activity of CAR-T cells in vitro and in vivo.
Collapse
Affiliation(s)
- Meng Zhang
- First Center Clinical College, Tianjin Medical University, Tianjin, 300192, China
| | - Xin Jin
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Rui Sun
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300192, China.,School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xia Xiong
- First Center Clinical College, Tianjin Medical University, Tianjin, 300192, China
| | - Jiaxi Wang
- First Center Clinical College, Tianjin Medical University, Tianjin, 300192, China
| | - Danni Xie
- First Center Clinical College, Tianjin Medical University, Tianjin, 300192, China
| | - MingFeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300192, China.
| |
Collapse
|
23
|
Vanhaesebroeck B, Perry MWD, Brown JR, André F, Okkenhaug K. PI3K inhibitors are finally coming of age. Nat Rev Drug Discov 2021; 20:741-769. [PMID: 34127844 PMCID: PMC9297732 DOI: 10.1038/s41573-021-00209-1] [Citation(s) in RCA: 226] [Impact Index Per Article: 75.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2021] [Indexed: 01/08/2023]
Abstract
Overactive phosphoinositide 3-kinase (PI3K) in cancer and immune dysregulation has spurred extensive efforts to develop therapeutic PI3K inhibitors. Although progress has been hampered by issues such as poor drug tolerance and drug resistance, several PI3K inhibitors have now received regulatory approval - the PI3Kα isoform-selective inhibitor alpelisib for the treatment of breast cancer and inhibitors mainly aimed at the leukocyte-enriched PI3Kδ in B cell malignancies. In addition to targeting cancer cell-intrinsic PI3K activity, emerging evidence highlights the potential of PI3K inhibitors in cancer immunotherapy. This Review summarizes key discoveries that aid the clinical translation of PI3Kα and PI3Kδ inhibitors, highlighting lessons learnt and future opportunities.
Collapse
Affiliation(s)
| | - Matthew W D Perry
- Medicinal Chemistry, Research and Early Development, Respiratory & Immunology BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jennifer R Brown
- CLL Center, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Fabrice André
- Institut Gustave Roussy, INSERM U981, Université Paris Saclay, Paris, France
| | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
24
|
Johansen KH, Golec DP, Thomsen JH, Schwartzberg PL, Okkenhaug K. PI3K in T Cell Adhesion and Trafficking. Front Immunol 2021; 12:708908. [PMID: 34421914 PMCID: PMC8377255 DOI: 10.3389/fimmu.2021.708908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
PI3K signalling is required for activation, differentiation, and trafficking of T cells. PI3Kδ, the dominant PI3K isoform in T cells, has been extensively characterised using PI3Kδ mutant mouse models and PI3K inhibitors. Furthermore, characterisation of patients with Activated PI3K Delta Syndrome (APDS) and mouse models with hyperactive PI3Kδ have shed light on how increased PI3Kδ activity affects T cell functions. An important function of PI3Kδ is that it acts downstream of TCR stimulation to activate the major T cell integrin, LFA-1, which controls transendothelial migration of T cells as well as their interaction with antigen-presenting cells. PI3Kδ also suppresses the cell surface expression of CD62L and CCR7 which controls the migration of T cells across high endothelial venules in the lymph nodes and S1PR1 which controls lymph node egress. Therefore, PI3Kδ can control both entry and exit of T cells from lymph nodes as well as the recruitment to and retention of T cells within inflamed tissues. This review will focus on the regulation of adhesion receptors by PI3Kδ and how this contributes to T cell trafficking and localisation. These findings are relevant for our understanding of how PI3Kδ inhibitors may affect T cell redistribution and function.
Collapse
Affiliation(s)
- Kristoffer H Johansen
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.,Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, United States
| | - Dominic P Golec
- Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, United States
| | - Julie H Thomsen
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | | | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
25
|
Li W, Lu L, Lu J, Wang X, Yang C, Jin J, Wu L, Hong X, Li F, Cao D, Yang Y, Wu M, Su B, Cheng J, Yang X, Di W, Deng L. cGAS-STING-mediated DNA sensing maintains CD8 + T cell stemness and promotes antitumor T cell therapy. Sci Transl Med 2021; 12:12/549/eaay9013. [PMID: 32581136 DOI: 10.1126/scitranslmed.aay9013] [Citation(s) in RCA: 128] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 02/03/2020] [Accepted: 05/28/2020] [Indexed: 12/13/2022]
Abstract
Although cGAS-STING-mediated DNA sensing in tumor cells or phagocytes is central for launching antitumor immunity, the role of intrinsic cGAS-STING activation in T cells remains unknown. Here, we observed that peripheral blood CD8+ T cells from patients with cancer showed remarkably compromised expression of the cGAS-STING cascade. We demonstrated that the cGAS-STING cascade in adoptively transferred CD8+ T cells was essential for antitumor immune responses in the context of T cell therapy in mice. Mechanistically, cell-autonomous cGAS and STING promoted the maintenance of stem cell-like CD8+ T cells, in part, by regulating the transcription factor TCF1 expression. Moreover, autocrine cGAS-STING-mediated type I interferon signaling augmented stem cell-like CD8+ T cell differentiation program mainly by restraining Akt activity. In addition, genomic DNA was selectively enriched in the cytosol of mouse CD8+ T cells upon in vitro and in vivo stimulation. STING agonism enhanced the formation of stem-like central memory CD8+ T cells from patients with cancer and potentiated antitumor responses of CAR-T cell therapy in a xenograft model. These findings advance our understanding of inherent cGAS-STING activation in T cells and provide insight into the development of improved T cell therapy by harnessing the cGAS-STING pathway for cancer immunotherapy.
Collapse
Affiliation(s)
- Wenwen Li
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital and Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lu Lu
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital and Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Juanjuan Lu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xinran Wang
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Chao Yang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jingsi Jin
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lingling Wu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaochuan Hong
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Fanlin Li
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Dongqing Cao
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yuanqin Yang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Meng Wu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Bing Su
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jinke Cheng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xuanming Yang
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Wen Di
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| | - Liufu Deng
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital and Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
26
|
Fan C, Lu W, Li K, Zhao C, Wang F, Ding G, Wang J. Identification of immune cell infiltration pattern and related critical genes in metastatic castration-resistant prostate cancer by bioinformatics analysis. Cancer Biomark 2021; 32:363-377. [PMID: 34151837 DOI: 10.3233/cbm-203222] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Metastatic castration-resistant prostate cancer (mCRPC) is the lethal stage of prostate cancer and the main cause of morbidity and mortality, which is also a potential target for immunotherapy. METHOD In this study, using the Approximate Relative Subset of RNA Transcripts (CIBERSORT) online method, we analysed the immune cell abundance ratio of each sample in the mCRPC dataset. The EdgeR (an R package) was used to classify differentially expressed genes (DEGs). Using the Database for annotation, visualisation and interactive exploration (DAVID) online method, we performed functional enrichment analyses. STRING online database and Cytoscape tools have been used to analyse protein-protein interaction (PPI) and classify hub genes. RESULTS The profiles of immune infiltration in mCRPC showed that Macrophages M2, Macrophages M0, T cells CD4 memory resting, T cells CD8 and Plasma cells were the main infiltration cell types in mCRPC samples. Macrophage M0 and T cell CD4 memory resting abundance ratios were correlated with clinical outcomes. We identified 1102 differentially expressed genes (DEGs) associated with the above two immune cells to further explore the underlying mechanisms. Enrichment analysis found that DEGs were substantially enriched in immune response, cell metastasis, and metabolism related categories. We identified 20 hub genes by the protein-protein interaction network analysis. Further analysis showed that three critical hub genes, CCR5, COL1A1 and CXCR3, were significantly associated with prostate cancer prognosis. CONCLUSION Our findings revealed the pattern of immune cell infiltration in mCRPC, and identified the types and genes of immune cells correlated with clinical outcomes. A new theoretical basis for immunotherapy may be given by our results.
Collapse
Affiliation(s)
- Caibin Fan
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei Lu
- School of Nursing, Suzhou Vocational Health and Technical College, Suzhou, Jiangsu, China.,Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kai Li
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chunchun Zhao
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fei Wang
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guanxiong Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianqing Wang
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
27
|
Huang J, Feng W, Li S, Tang H, Qin S, Li W, Gong Y, Fang Y, Liu Y, Wang S, Guo Y, Xu Z, Shen Q. Berberine Exerts Anti-cancer Activity by Modulating Adenosine Monophosphate- Activated Protein Kinase (AMPK) and the Phosphatidylinositol 3-Kinase/ Protein Kinase B (PI3K/AKT) Signaling Pathways. Curr Pharm Des 2021; 27:565-574. [PMID: 32988344 DOI: 10.2174/1381612826666200928155728] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/12/2020] [Indexed: 11/22/2022]
Abstract
Background The antagonistic relationship between adenosine monophosphate-activated protein kinase (AMPK) and phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) signaling play a vital role in cancer development. The anti-cancer effects of berberine have been reported as a main component of the traditional Chinese medicine Rhizoma coptidis, although the roles of these signaling pathways in these effects have not been systematically reviewed. METHODS We searched the PubMed database for studies with keywords including ["berberine"] and ["tumor" or "cancer"] and ["AMPK"] or ["AKT"] published between January 2010 and July 2020, to elucidate the roles of the AMPK and PI3K/AKT pathways and their upstream and downstream targets in the anti-cancer effects of berberine. RESULTS The anti-cancer effects of berberine include inhibition of cancer cell proliferation, promotion of apoptosis and autophagy in cancer cells, and prevention of metastasis and angiogenesis. The mechanism of these effects involves multiple cell kinases and signaling pathways, including activation of AMPK and forkhead box transcription factor O3a (FOXO3a), accumulation of reactive oxygen species (ROS), and inhibition of the activity of PI3K/AKT, rapamycin (mTOR) and nuclear factor-κB (NF-κB). Most of these mechanisms converge on regulation of the balance of AMPK and PI3K/AKT signaling by berberine. CONCLUSION This evidence supports the possibility that berberine is a promising anti-cancer natural product, with pharmaceutical potential in inhibiting cancer growth, metastasis and angiogenesis via multiple pathways, particularly by regulating the balance of AMPK and PI3K/AKT signaling. However, systematic preclinical studies are still required to provide scientific evidence for further clinical studies.
Collapse
Affiliation(s)
- Jin Huang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Wei Feng
- Emergercy Department, South Branch of Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing100053, China
| | - Shanshan Li
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Huiling Tang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Siru Qin
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Wei Li
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yinan Gong
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuxin Fang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yangyang Liu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shenjun Wang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yi Guo
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhifang Xu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qian Shen
- Department of Massage and Physiotherapy, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, China
| |
Collapse
|
28
|
Dong C, Wu J, Chen Y, Nie J, Chen C. Activation of PI3K/AKT/mTOR Pathway Causes Drug Resistance in Breast Cancer. Front Pharmacol 2021; 12:628690. [PMID: 33790792 PMCID: PMC8005514 DOI: 10.3389/fphar.2021.628690] [Citation(s) in RCA: 156] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/18/2021] [Indexed: 12/16/2022] Open
Abstract
Although chemotherapy, targeted therapy and endocrine therapy decrease rate of disease recurrence in most breast cancer patients, many patients exhibit acquired resistance. Hyperactivation of the PI3K/AKT/mTOR pathway is associated with drug resistance and cancer progression. Currently, a number of drugs targeting PI3K/AKT/mTOR are being investigated in clinical trials by combining them with standard therapies to overcome acquired resistance in breast cancer. In this review, we summarize the critical role of the PI3K/AKT/mTOR pathway in drug resistance, the development of PI3K/AKT/mTOR inhibitors, and strategies to overcome acquired resistance to standard therapies in breast cancer.
Collapse
Affiliation(s)
- Chao Dong
- Department of the Second Medical Oncology, The 3rd Affiliated Hospital of Kunming Medical University, Yunnan Tumor Hospital, Kunming, China
| | - Jiao Wu
- Department of the Second Medical Oncology, The 3rd Affiliated Hospital of Kunming Medical University, Yunnan Tumor Hospital, Kunming, China
| | - Yin Chen
- Department of Urology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Jianyun Nie
- Department of the Third Breast Surgery, The 3rd Affiliated Hospital of Kunming Medical University, Yunnan Tumor Hospital, Kunming, China
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
29
|
Petroni G, Buqué A, Zitvogel L, Kroemer G, Galluzzi L. Immunomodulation by targeted anticancer agents. Cancer Cell 2021; 39:310-345. [PMID: 33338426 DOI: 10.1016/j.ccell.2020.11.009] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/17/2020] [Accepted: 11/17/2020] [Indexed: 02/08/2023]
Abstract
At odds with conventional chemotherapeutics, targeted anticancer agents are designed to inhibit precise molecular alterations that support oncogenesis or tumor progression. Despite such an elevated degree of molecular specificity, many clinically employed and experimental targeted anticancer agents also mediate immunostimulatory or immunosuppressive effects that (at least in some settings) influence therapeutic efficacy. Here, we discuss the main immunomodulatory effects of targeted anticancer agents and explore potential avenues to harness them in support of superior clinical efficacy.
Collapse
Affiliation(s)
- Giulia Petroni
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Aitziber Buqué
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Laurence Zitvogel
- Gustave Roussy Cancer Center, Villejuif, France; INSERM U1015, Villejuif, France; Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France; Faculty of Medicine, Paris-Saclay University, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Equipe Labellisée Par La Ligue Contre le Cancer, Centre de Recherche des Cordeliers, INSERM U1138, Université de Paris, Sorbonne Université, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China; Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA; Department of Dermatology, Yale School of Medicine, New Haven, CT, USA; Université de Paris, Paris, France.
| |
Collapse
|
30
|
T-Cell Dysfunction as a Limitation of Adoptive Immunotherapy: Current Concepts and Mitigation Strategies. Cancers (Basel) 2021; 13:cancers13040598. [PMID: 33546277 PMCID: PMC7913380 DOI: 10.3390/cancers13040598] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/29/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary T cells are immune cells that can be used to target infections or cancers. Adoptive T-cell immunotherapy leverages these properties and/or confers new features to T cells through ex vivo manipulations prior to their use in patients. However, as a “living drug,” the function of these cells can be hampered by several built-in physiological constraints and external factors that limit their efficacy. Manipulating T cells ex vivo can impart dysfunctional features to T cells through repeated stimulations and expansion, but it also offers many opportunities to improve the therapeutic potential of these cells, including emerging interventions to prevent or reverse T-cell dysfunction developing ex vivo or after transfer in patients. This review outlines the various forms of T-cell dysfunction, emphasizes how it affects various types of T-cell immunotherapy approaches, and describes current and anticipated strategies to limit T-cell dysfunction. Abstract Over the last decades, cellular immunotherapy has revealed its curative potential. However, inherent physiological characteristics of immune cells can limit the potency of this approach. Best defined in T cells, dysfunction associated with terminal differentiation, exhaustion, senescence, and activation-induced cell death, undermine adoptive cell therapies. In this review, we concentrate on how the multiple mechanisms that articulate the various forms of immune dysfunction impact cellular therapies primarily involving conventional T cells, but also other lymphoid subtypes. The repercussions of immune cell dysfunction across the full life cycle of cell therapy, from the source material, during manufacturing, and after adoptive transfer, are discussed, with an emphasis on strategies used during ex vivo manipulations to limit T-cell dysfunction. Applicable to cellular products prepared from native and unmodified immune cells, as well as genetically engineered therapeutics, the understanding and potential modulation of dysfunctional features are key to the development of improved cellular immunotherapies.
Collapse
|
31
|
Rostamian H, Fallah-Mehrjardi K, Khakpoor-Koosheh M, Pawelek JM, Hadjati J, Brown CE, Mirzaei HR. A metabolic switch to memory CAR T cells: Implications for cancer treatment. Cancer Lett 2020; 500:107-118. [PMID: 33290868 DOI: 10.1016/j.canlet.2020.12.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/27/2022]
Abstract
Therapeutic efficacy of chimeric antigen receptor (CAR) T cells is associated with their expansion, persistence and effector function. Although CAR T cell therapy has shown remarkable therapeutic effects in hematological malignancies, its therapeutic efficacy has been limited in some types of cancers - in particular, solid tumors - partially due to the cells' inability to persist and the acquisition of T cell dysfunction within a harsh immunosuppressive tumor microenvironment. Therefore, it would be expected that generation of CAR T cells with intrinsic properties for functional longevity, such as the cells with early-memory phenotypes, could beneficially enhance antitumor immunity. Furthermore, because the metabolic pathways of CAR T cells help determine cellular differentiation and lifespan, therapies targeting such pathways like glycolysis and oxidative phosphorylation, can alter CAR T cell fate and durability within tumors. Here we discuss how reprogramming of CAR T cell metabolism and metabolic switch to memory CAR T cells influences their antitumor activity. We also offer potential strategies for targeting these metabolic circuits in the setting of adoptive CAR T cell therapy, aiming to better unleash the potential of adoptive CAR T cell therapy in the clinic.
Collapse
Affiliation(s)
- Hosein Rostamian
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Keyvan Fallah-Mehrjardi
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Khakpoor-Koosheh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - John M Pawelek
- Department of Dermatology and the Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Jamshid Hadjati
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Christine E Brown
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, CA, 91010, USA; Department of Immuno-Oncology, City of Hope Beckman Research Institute, Duarte, CA, 91010, USA.
| | - Hamid R Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
32
|
Cell composition and expansion strategy can reduce the beneficial effect of AKT-inhibition on functionality of CD8 + T cells. Cancer Immunol Immunother 2020; 69:2259-2273. [PMID: 32504246 PMCID: PMC7568704 DOI: 10.1007/s00262-020-02612-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 05/15/2020] [Indexed: 12/29/2022]
Abstract
AKT-inhibition is a promising approach to improve T cell therapies; however, its effect on CD4+ T cells is insufficiently explored. Previously, we and others showed that AKT-inhibition during ex vivo CD8+ T cell expansion facilitates the generation of polyfunctional T cells with stem cell memory-like traits. However, most therapeutic T cell products are generated from lymphocytes, containing CD4+ T cells that can affect CD8+ T cells dependent on the Th-subset. Here, we investigated the effect of AKT-inhibition on CD4+ T cells, during separate as well as total T cell expansions. Interestingly, ex vivo AKT-inhibition preserved the early memory phenotype of CD4+ T cells based on higher CD62L, CXCR4 and CCR7 expression. However, in the presence of AKT-inhibition, Th-differentiation was skewed toward more Th2-associated at the expense of Th1-associated cells. Importantly, the favorable effect of AKT-inhibition on the functionality of CD8+ T cells drastically diminished in the presence of CD4+ T cells. Moreover, also the expansion method influenced the effect of AKT-inhibition on CD8+ T cells. These findings indicate that the effect of AKT-inhibition on CD8+ T cells is dependent on cell composition and expansion strategy, where presence of CD4+ T cells as well as polyclonal stimulation impede the favorable effect of AKT-inhibition.
Collapse
|
33
|
Wang J, Hasan F, Frey AC, Li HS, Park J, Pan K, Haymaker C, Bernatchez C, Lee DA, Watowich SS, Yee C. Histone Deacetylase Inhibitors and IL21 Cooperate to Reprogram Human Effector CD8 + T Cells to Memory T Cells. Cancer Immunol Res 2020; 8:794-805. [PMID: 32213626 PMCID: PMC7269845 DOI: 10.1158/2326-6066.cir-19-0619] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 12/27/2019] [Accepted: 03/17/2020] [Indexed: 12/18/2022]
Abstract
Clinical response rates after adoptive cell therapy (ACT) are highly correlated with in vivo persistence of the infused T cells. However, antigen-specific T cells found in tumor sites are often well-differentiated effector cells with limited persistence. Central memory CD8+ T cells, capable of self-renewal, represent desirable ACT products. We report here that exposure to a histone deacetylase inhibitor (HDACi) and IL21 could reprogram differentiated human CD8+ T cells into central memory-like T cells. Dedifferentiation of CD8+ T cells was initiated by increased H3 acetylation and chromatin accessibility at the CD28 promoter region. This led to IL21-mediated pSTAT3 binding to the CD28 region, and subsequent upregulation of surface CD28 and CD62L (markers of central memory T cells). The reprogrammed cells exhibited enhanced proliferation in response to both IL2 and IL15, and a stable memory-associated transcriptional signature (increased Lef1 and Tcf7). Our findings support the application of IL21 and HDACi for the in vitro generation of highly persistent T-cell populations that can augment the efficacy of adoptively transferred T cells.
Collapse
Affiliation(s)
- Junmei Wang
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Farah Hasan
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amanda C Frey
- Molecular and Cellular Biology Program, University of Washington, Seattle, Washington
| | - Haiyan S Li
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jungsun Park
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ke Pan
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cara Haymaker
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chantale Bernatchez
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dean A Lee
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stephanie S Watowich
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cassian Yee
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
34
|
Guedan S, Madar A, Casado-Medrano V, Shaw C, Wing A, Liu F, Young RM, June CH, Posey AD. Single residue in CD28-costimulated CAR-T cells limits long-term persistence and antitumor durability. J Clin Invest 2020; 130:3087-3097. [PMID: 32069268 PMCID: PMC7260017 DOI: 10.1172/jci133215] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 02/13/2020] [Indexed: 12/21/2022] Open
Abstract
Chimeric antigen receptor-T (CAR-T) cell therapies can eliminate relapsed and refractory tumors, but the durability of antitumor activity requires in vivo persistence. Differential signaling through the CAR costimulatory domain can alter the T cell metabolism, memory differentiation, and influence long-term persistence. CAR-T cells costimulated with 4-1BB or ICOS persist in xenograft models but those constructed with CD28 exhibit rapid clearance. Here, we show that a single amino acid residue in CD28 drove T cell exhaustion and hindered the persistence of CD28-based CAR-T cells and changing this asparagine to phenylalanine (CD28-YMFM) promoted durable antitumor control. In addition, CD28-YMFM CAR-T cells exhibited reduced T cell differentiation and exhaustion as well as increased skewing toward Th17 cells. Reciprocal modification of ICOS-containing CAR-T cells abolished in vivo persistence and antitumor activity. This finding suggests modifications to the costimulatory domains of CAR-T cells can enable longer persistence and thereby improve antitumor response.
Collapse
Affiliation(s)
- Sonia Guedan
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Hematology, Hospital Clinic, Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Aviv Madar
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| | - Victoria Casado-Medrano
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Carolyn Shaw
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anna Wing
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Fang Liu
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Regina M. Young
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Carl H. June
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine and
- Parker Institute for Cellular Immunotherapy, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Avery D. Posey
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine and
- Parker Institute for Cellular Immunotherapy, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
35
|
Janelle V, Rulleau C, Del Testa S, Carli C, Delisle JS. T-Cell Immunotherapies Targeting Histocompatibility and Tumor Antigens in Hematological Malignancies. Front Immunol 2020; 11:276. [PMID: 32153583 PMCID: PMC7046834 DOI: 10.3389/fimmu.2020.00276] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/03/2020] [Indexed: 12/19/2022] Open
Abstract
Over the last decades, T-cell immunotherapy has revealed itself as a powerful, and often curative, strategy to treat blood cancers. In hematopoietic cell transplantation, most of the so-called graft-vs.-leukemia (GVL) effect hinges on the recognition of histocompatibility antigens that reflect immunologically relevant genetic variants between donors and recipients. Whether other variants acquired during the neoplastic transformation, or the aberrant expression of gene products can yield antigenic targets of similar relevance as the minor histocompatibility antigens is actively being pursued. Modern genomics and proteomics have enabled the high throughput identification of candidate antigens for immunotherapy in both autologous and allogeneic settings. As such, these major histocompatibility complex-associated tumor-specific (TSA) and tumor-associated antigens (TAA) can allow for the targeting of multiple blood neoplasms, which is a limitation for other immunotherapeutic approaches, such as chimeric antigen receptor (CAR)-modified T cells. We review the current strategies taken to translate these discoveries into T-cell therapies and propose how these could be introduced in clinical practice. Specifically, we discuss the criteria that are used to select the antigens with the greatest therapeutic value and we review the various T-cell manufacturing approaches in place to either expand antigen-specific T cells from the native repertoire or genetically engineer T cells with minor histocompatibility antigen or TSA/TAA-specific recombinant T-cell receptors. Finally, we elaborate on the current and future incorporation of these therapeutic T-cell products into the treatment of hematological malignancies.
Collapse
Affiliation(s)
- Valérie Janelle
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| | - Caroline Rulleau
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| | - Simon Del Testa
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| | - Cédric Carli
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| | - Jean-Sébastien Delisle
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Médecine, Université de Montréal, Montréal, QC, Canada.,Division Hématologie et Oncologie, Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| |
Collapse
|
36
|
Cao J, Yang X, Li J, Wu H, Li P, Yao Z, Dong Z, Tian J. Screening and Identifying Immune-Related Cells and Genes in the Tumor Microenvironment of Bladder Urothelial Carcinoma: Based on TCGA Database and Bioinformatics. Front Oncol 2020; 9:1533. [PMID: 32010623 PMCID: PMC6974676 DOI: 10.3389/fonc.2019.01533] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022] Open
Abstract
Bladder cancer is the most common cancer of the urinary system and its treatment has scarcely progressed for nearly 30 years. Advances in checkpoint inhibitor research have seemingly provided a new approach for treatment. However, there have been issues predicting immunotherapeutic biomarkers and identifying new therapeutic targets. We downloaded the gene expression profile and clinical data of 408 cases bladder urinary cancer from the Cancer Genome Atlas (TCGA) portal, and the abundance ratio of immune cells for each sample was obtained via the "Cell Type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT)" algorithm. Then, four survival-related immune cells were obtained via Kaplan-Meier survival analysis, and 933 immune-related genes were obtained via a variance analysis. Enrichment, protein-protein interaction, and co-expression analyses were performed for these genes. Lastly, 4 survival-related immune cells and 24 hub genes were identified, four of which were related to overall survival. More importantly, these immune cells and genes were closely related to the clinical features. These cells and genes may have research value and clinical application in bladder cancer immunotherapy. Our study not only provides cell and gene targets for bladder cancer immunotherapy, but also provides new ideas for researchers to explore the immunotherapy of various tumors.
Collapse
Affiliation(s)
- Jinlong Cao
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Urological Diseases of Gansu Provincial, Lanzhou, China
| | - Xin Yang
- Reproductive Medicine Center, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Jianpeng Li
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Urological Diseases of Gansu Provincial, Lanzhou, China
| | - Hao Wu
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Urological Diseases of Gansu Provincial, Lanzhou, China
| | - Pan Li
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Urological Diseases of Gansu Provincial, Lanzhou, China
| | - Zhiqiang Yao
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Urological Diseases of Gansu Provincial, Lanzhou, China
| | - Zhichun Dong
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Urological Diseases of Gansu Provincial, Lanzhou, China
| | - Junqiang Tian
- Key Laboratory of Urological Diseases of Gansu Provincial, Lanzhou, China
| |
Collapse
|
37
|
Abstract
As a specifically programmable, living immunotherapeutic drug, chimeric antigen receptor (CAR)-modified T cells are providing an alternative treatment option for a broad variety of diseases including so far refractory cancer. By recognizing a tumor-associated antigen, the CAR triggers an anti-tumor response of engineered patient's T cells achieving lasting remissions in the treatment of leukemia and lymphoma. During the last years, significant progress was made in optimizing the CAR design, in manufacturing CAR-engineered T cells, and in the clinical management of patients showing promise to establish adoptive CAR T cell therapy as an effective treatment option in the forefront.
Collapse
Affiliation(s)
- Astrid Holzinger
- RCI Regensburg Center for Interventional Immunology, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany
- Chair Genetic Immunotherapy, RCI c/o University Hospital Regensburg, Regensburg, Germany
| | - Hinrich Abken
- RCI Regensburg Center for Interventional Immunology, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany.
- Chair Genetic Immunotherapy, RCI c/o University Hospital Regensburg, Regensburg, Germany.
| |
Collapse
|
38
|
Abstract
CD8+T cells are important in protective immunity against intracellular pathogens and tumors. In chronic infections or cancer, CD8+T cells are constantly exposed to antigens and inflammatory signals. Such excessive and constitutive signals lead to the deterioration of T cell function, called 'exhaustion'. Exhausted T cells are characterized by low proliferation in response to antigen stimulation, progressive loss of effector function (cytokine production and killing function), expression of multiple inhibitory receptors such as PD-1, Tim3, and LAG3, and metabolic alterations from oxidative phosphorylation to glycolysis. These dysfunctions are associated with altered transcriptional programs and epigenetic regulations and recent studies suggested that NR4a and TOX transcription factors are deeply involved in exhaustion phenotypes. However, an increase the early memory T cells including stem cell memory T (TSCM) cells is critical for T cell persistence and efficient tumor killing especially for adoptive cancer immunotherapy such as CAR-T cell therapy. An increasing amount of evidence supports the therapeutic potential of targeting exhausted T cells and TSCM cells. We have begun to understand the molecular mechanisms of T cell exhaustion and early memory formation, and the clinical application of converting exhausted T cells to rejuvenated early memory T cells is the goal of our study.
Collapse
Affiliation(s)
- Makoto Ando
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Minako Ito
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Tanakorn Srirat
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Taisuke Kondo
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
39
|
Pilipow K, Scamardella E, Lugli E. Generating stem-like memory T cells with antioxidants for adoptive cell transfer immunotherapy of cancer. Methods Enzymol 2019; 631:137-158. [PMID: 31948545 DOI: 10.1016/bs.mie.2019.08.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Among the multiple factors that are responsible for the success of adoptive cell transfer (ACT) immunotherapy for cancer, the differentiation status of the in vitro expanded T cell product at the time of transfer seems to play a major role. In particular, less differentiated memory CD8+ T cells endowed with self-renewing capacity and multipotency exert the most potent antitumor activity. To this aim, expansion protocols that generate sufficient numbers of tumor-specific CD8+ T cells with superior capacity to persist in vivo following ACT are needed. We describe a procedure for the differentiation of TCF-1+ stem-like CD8+ memory T cells from peripheral blood naïve precursors that takes advantage of the use of antioxidants, in particular N-acetylcysteine (NAC), in combination with T cell receptor stimulation and proinflammatory cytokines. We additionally describe how to conduct in vitro assays to test the stem-like features of the generated cells at the phenotypic, functional and metabolic level. Balancing the oxidative metabolism by the addition of antioxidants during in vitro manipulation of CD8+ T cells results in the generation of cell products with potent antitumor characteristics following ACT.
Collapse
Affiliation(s)
- Karolina Pilipow
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Eloise Scamardella
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Enrico Lugli
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy; Flow Cytometry Core, Humanitas Clinical and Research Center, Rozzano, Milan, Italy.
| |
Collapse
|
40
|
Revising PTEN in the Era of Immunotherapy: New Perspectives for an Old Story. Cancers (Basel) 2019; 11:cancers11101525. [PMID: 31658667 PMCID: PMC6826982 DOI: 10.3390/cancers11101525] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/17/2019] [Accepted: 09/30/2019] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy has emerged as the new therapeutic frontier of cancer treatment, showing enormous survival benefits in multiple tumor diseases. Although undeniable success has been observed in clinical trials, not all patients respond to treatment. Different concurrent conditions can attenuate or completely abrogate the usefulness of immunotherapy due to the activation of several escape mechanisms. Indeed, the tumor microenvironment has an almost full immunosuppressive profile, creating an obstacle to therapeutic treatment. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) governs a plethora of cellular processes, including maintenance of genomic stability, cell survival/apoptosis, migration, and metabolism. The repertoire of PTEN functions has recently been expanded to include regulation of the tumor microenvironment and immune system, leading to a drastic reevaluation of the canonical paradigm of PTEN action with new potential implications for immunotherapy-based approaches. Understanding the implication of PTEN in cancer immunoediting and immune evasion is crucial to develop new cancer intervention strategies. Recent evidence has shown a double context-dependent role of PTEN in anticancer immunity. Here we summarize the current knowledge of PTEN’s role at a crossroads between tumor and immune compartments, highlighting the most recent findings that are likely to change future clinical practice.
Collapse
|
41
|
Page DB, Bear H, Prabhakaran S, Gatti-Mays ME, Thomas A, Cobain E, McArthur H, Balko JM, Gameiro SR, Nanda R, Gulley JL, Kalinsky K, White J, Litton J, Chmura SJ, Polley MY, Vincent B, Cescon DW, Disis ML, Sparano JA, Mittendorf EA, Adams S. Two may be better than one: PD-1/PD-L1 blockade combination approaches in metastatic breast cancer. NPJ Breast Cancer 2019; 5:34. [PMID: 31602395 PMCID: PMC6783471 DOI: 10.1038/s41523-019-0130-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/05/2019] [Indexed: 01/07/2023] Open
Abstract
Antibodies blocking programmed death 1 (anti-PD-1) or its ligand (anti-PD-L1) are associated with modest response rates as monotherapy in metastatic breast cancer, but are generally well tolerated and capable of generating dramatic and durable benefit in a minority of patients. Anti-PD-1/L1 antibodies are also safe when administered in combination with a variety of systemic therapies (chemotherapy, targeted therapies), as well as with radiotherapy. We summarize preclinical, translational, and preliminary clinical data in support of combination approaches with anti-PD-1/L1 in metastatic breast cancer, focusing on potential mechanisms of synergy, and considerations for clinical practice and future investigation.
Collapse
Affiliation(s)
- David B. Page
- Providence Cancer Institute; Earle A. Chiles Research Institute, Portland, OR USA
| | - Harry Bear
- Division of Surgical Oncology and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA USA
| | - Sangeetha Prabhakaran
- Department of Surgery, Division of Surgery, University of New Mexico; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM USA
| | | | - Alexandra Thomas
- Wake Forest University School of Medicine, Winston-Salem, NC USA
| | | | | | - Justin M. Balko
- Department of Medicine and Breast Cancer Research Program, Vanderbilt University Medical Center, Nashville, TN USA
| | - Sofia R. Gameiro
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, Bethesda, MD USA
| | - Rita Nanda
- The University of Chicago, Chicago, IL USA
| | - James L. Gulley
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| | | | - Julia White
- Ohio State Wexner Medical Center, Columbus, OH USA
| | | | | | | | | | - David W. Cescon
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON Canada
| | | | - Joseph A. Sparano
- Department of Medical Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY USA
| | - Elizabeth A. Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women’s Hospital; Breast Oncology Program, Dana-Farber/Brigham and Women’s Cancer Center, Boston, MA USA
| | - Sylvia Adams
- Perlmutter Cancer Center, NYU School of Medicine, New York, NY USA
| |
Collapse
|
42
|
Jiang X, Xu J, Liu M, Xing H, Wang Z, Huang L, Mellor AL, Wang W, Wu S. Adoptive CD8 + T cell therapy against cancer:Challenges and opportunities. Cancer Lett 2019; 462:23-32. [PMID: 31356845 DOI: 10.1016/j.canlet.2019.07.017] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/11/2019] [Accepted: 07/23/2019] [Indexed: 12/12/2022]
Abstract
Cancer immunotherapy is a new and promising option for cancer treatment. Unlike traditional chemo- and radiotherapy, immunotherapy actives host immune system to attack malignancies, and this potentially offers long-term protection from recurrence with less toxicity in comparison to conventional chemo- and radiation therapy. In adoptive CD8+ T cell therapy (ACT), large numbers of tumor-specific T cells are sourced from patients and expanded in vitro and infused back to patients. T cells can be expanded from naturally-induced tumor-specific CD8+ T cells isolated from tumor infiltrating lymphocytes (TIL) or genetically-modified autologous circulating CD8+ T cells. The engineered T cells expressed tumor-specific antigen receptors including chimeric antigen receptors (CARs) and T cell receptors (TCRs), prepared from cultured B and T cell clones, respectively. The most successful ACT, anti-CD19 chimeric antigen receptor T (CAR-T) cell therapy directed against B cell lymphoma, is already approved for use based on evidence of efficacy. Efficacy of solid tumors is not yet forthcoming. This review summarizes current technology developments using ACT in clinical trials. In this review, differences between various ACT approaches are discussed. Furthermore, resistance factors in the tumor microenvironment are also considered, as are immune related adverse effects, critical clinic monitoring parameters and potential mitigation approaches.
Collapse
Affiliation(s)
- Xiaotao Jiang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangdong Provincial Key Laboratory of Proteomics, Guangzhou, Guangdong, People's Republic of China.
| | - Jiang Xu
- Department of Rehabilitation, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, People's Republic of China
| | - Mingfeng Liu
- Department of Breast, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| | - Hui Xing
- Department of Obstetrics and Gynecology, Xiangyang Central Hospital, Xiangyang, Hubei, People's Republic of China.
| | - Zhiming Wang
- Sino-British Research Center for Molecular Oncology, National Center for the International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, People's Republic of China.
| | - Lei Huang
- Institute of Cellular Medicine, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle-Upon-Tyne, United Kingdom.
| | - Andrew L Mellor
- Institute of Cellular Medicine, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle-Upon-Tyne, United Kingdom.
| | - Wei Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China.
| | - Sha Wu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangdong Provincial Key Laboratory of Proteomics, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
43
|
Dahmani A, Janelle V, Carli C, Richaud M, Lamarche C, Khalili M, Goupil M, Bezverbnaya K, Bramson JL, Delisle JS. TGFβ Programs Central Memory Differentiation in Ex Vivo-Stimulated Human T Cells. Cancer Immunol Res 2019; 7:1426-1439. [PMID: 31308016 DOI: 10.1158/2326-6066.cir-18-0691] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 03/27/2019] [Accepted: 07/09/2019] [Indexed: 11/16/2022]
Abstract
The adoptive transfer of ex vivo-expanded T cells is a promising approach to treat several malignancies. Several lines of evidence support that the infusion of T cells with early memory features, capable of expanding and persisting after transfer, are associated with better outcomes. We report herein that exposure to exogenous TGFβ during human T-cell stimulation ex vivo leads to the accumulation of early/central memory (Tcm) cells. Exposure to TGFβ suppressed the expression of BLIMP-1, a key orchestrator of effector T-cell differentiation, and led to the upregulation of the memory-associated transcription factor ID3. Accordingly, this was associated with an early memory transcriptional signature in both CD4+ and CD8+ T-cell subsets. The T cells stimulated in the presence of TGFβ expanded normally, and displayed polyfunctional features and no suppressive activity. The adoptive transfer of ex vivo-stimulated T cells into immunodeficient mice confirmed that TGFβ-conditioned cells had an enhanced capacity to persist and mediate xenogeneic graft-versus-host disease, as predicted by their early T-cell memory phenotype. Chimeric antigen receptor-expressing T cells generated in the presence of exogenous TGFβ were cytotoxic and more effective at controlling tumor growth in immunodeficient animals. This work unveils a new role for TGFβ in memory T-cell differentiation and indicates that TGFβ signaling may be harnessed to program Tcm differentiation in the context of ex vivo T-cell stimulation for adoptive immunotherapy in humans.
Collapse
Affiliation(s)
- Amina Dahmani
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
| | - Valérie Janelle
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
| | - Cédric Carli
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
| | - Manon Richaud
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
| | - Caroline Lamarche
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Myriam Khalili
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
| | - Mathieu Goupil
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
| | - Ksenia Bezverbnaya
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan L Bramson
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jean-Sébastien Delisle
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada.
- Département de Médecine, Université de Montréal, Montreal, Quebec, Canada
- Hematology-Oncology Division, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| |
Collapse
|
44
|
Magalhaes I, Carvalho-Queiroz C, Hartana CA, Kaiser A, Lukic A, Mints M, Nilsson O, Grönlund H, Mattsson J, Berglund S. Facing the future: challenges and opportunities in adoptive T cell therapy in cancer. Expert Opin Biol Ther 2019; 19:811-827. [PMID: 30986360 DOI: 10.1080/14712598.2019.1608179] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION In recent years, immunotherapy for the treatment of solid cancer has emerged as a promising therapeutic alternative. Adoptive cell therapy (ACT), especially T cell-based, has been found to cause tumor regression and even cure in a percentage of treated patients. Checkpoint inhibitors further underscore the potential of the T cell compartment in the treatment of cancer. Not all patients respond to these treatments; however, many challenges remain. AREAS COVERED This review covers the challenges and progress in tumor antigen target identification and selection, and cell product manufacturing for T cell ACT. Tumor immune escape mechanisms and strategies to overcome those in the context of T cell ACT are also discussed. EXPERT OPINION The immunotherapy toolbox is rapidly expanding and improving, and the future promises further breakthroughs in the T cell ACT field. The heterogeneity of the tumor microenvironment and the multiplicity of tumor immune escape mechanisms pose formidable challenges to successful T cell immunotherapy in solid tumors, however. Individualized approaches and strategies combining treatments targeting different immunotherapeutic aspects will be needed in order to expand the applicability and improve the response rates in future.
Collapse
Affiliation(s)
- Isabelle Magalhaes
- a Department of Oncology-Pathology , Karolinska Institutet , Stockholm , Sweden
| | - Claudia Carvalho-Queiroz
- b Therapeutic Immune Design, Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden
| | - Ciputra Adijaya Hartana
- c Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital , Cambridge , MA , USA
| | - Andreas Kaiser
- b Therapeutic Immune Design, Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden
| | - Ana Lukic
- b Therapeutic Immune Design, Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden
| | - Michael Mints
- a Department of Oncology-Pathology , Karolinska Institutet , Stockholm , Sweden.,d Department of Surgical and Perioperative Sciences , Umeå University, Umeå, Sweden.,e Blood and Marrow Transplant Program, Medical Oncology and Hematology , Princess Margaret Cancer Center , Toronto , Canada.,f Department of Medicine , University of Toronto , Toronto , Canada
| | - Ola Nilsson
- b Therapeutic Immune Design, Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden
| | - Hans Grönlund
- b Therapeutic Immune Design, Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden
| | - Jonas Mattsson
- a Department of Oncology-Pathology , Karolinska Institutet , Stockholm , Sweden.,f Department of Medicine , University of Toronto , Toronto , Canada
| | - Sofia Berglund
- a Department of Oncology-Pathology , Karolinska Institutet , Stockholm , Sweden.,b Therapeutic Immune Design, Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden
| |
Collapse
|
45
|
Abstract
Genetically engineered T cells are powerful new medicines, offering hope for curative responses in patients with cancer. Chimeric antigen receptor (CAR) T cells were recently approved by the US Food and Drug Administration and are poised to enter the practice of medicine for leukemia and lymphoma, demonstrating that engineered immune cells can serve as a powerful new class of cancer therapeutics. The emergence of synthetic biology approaches for cellular engineering provides a broadly expanded set of tools for programming immune cells for enhanced function. Advances in T cell engineering, genetic editing, the selection of optimal lymphocytes, and cell manufacturing have the potential to broaden T cell-based therapies and foster new applications beyond oncology, in infectious diseases, organ transplantation, and autoimmunity.
Collapse
Affiliation(s)
- Sonia Guedan
- Department of Hematology, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| | - Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Parker Institute for Cellular Immunotherapy at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Parker Institute for Cellular Immunotherapy at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
46
|
Sarkar I, Pati S, Dutta A, Basak U, Sa G. T-memory cells against cancer: Remembering the enemy. Cell Immunol 2019; 338:27-31. [PMID: 30928016 DOI: 10.1016/j.cellimm.2019.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 03/14/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Recently various types of immunotherapies have made immense progress in combating cancer. Adoptive cell therapy, being one of the most favorable forms of immunotherapy, is rapidly moving from bench to bed. MAIN BODY Different types of T-memory cells are being used as promising candidates for adoptive cell therapy: T effector memory (TEM) cells which are terminally differentiated memory cells and attain effector function soon after re-stimulation; T central memory (TCM) cells which differentiate into effector T-memory subsets and T-effector cells after antigenic stimulation; and tissue T resident memory (TRM) cells which fight the tumor insult at the peripheral tissues. Recently, a new subtype of T-memory cells, T stem cell memory (TSCM) have been identified as the most favorable candidate for adoptive cell therapy as they exhibit higher persistence, anti-tumor immunity and self-renewal capacity in the tumor-bearing host. CONCLUSION In this review, we briefly describe the concept and types of T-memory cells as well as their role as potential candidates for anti-cancer immunotherapy.
Collapse
Affiliation(s)
- Irene Sarkar
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Subhadip Pati
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Abhishek Dutta
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Udit Basak
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Gaurisankar Sa
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India.
| |
Collapse
|
47
|
Carnevalli LS, Sinclair C, Taylor MA, Gutierrez PM, Langdon S, Coenen-Stass AML, Mooney L, Hughes A, Jarvis L, Staniszewska A, Crafter C, Sidders B, Hardaker E, Hudson K, Barry ST. PI3Kα/δ inhibition promotes anti-tumor immunity through direct enhancement of effector CD8 + T-cell activity. J Immunother Cancer 2018; 6:158. [PMID: 30587236 PMCID: PMC6307194 DOI: 10.1186/s40425-018-0457-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 11/20/2018] [Indexed: 01/08/2023] Open
Abstract
PI3K inhibitors with differential selectivity to distinct PI3K isoforms have been tested extensively in clinical trials, largely to target tumor epithelial cells. PI3K signaling also regulates the immune system and inhibition of PI3Kδ modulate the tumor immune microenvironment of pre-clinical mouse tumor models by relieving T-regs-mediated immunosuppression. PI3K inhibitors as a class and PI3Kδ specifically are associated with immune-related side effects. However, the impact of mixed PI3K inhibitors in tumor immunology is under-explored. Here we examine the differential effects of AZD8835, a dual PI3Kα/δ inhibitor, specifically on the tumor immune microenvironment using syngeneic models. Continuous suppression of PI3Kα/δ was not required for anti-tumor activity, as tumor growth inhibition was potentiated by an intermittent dosing/schedule in vivo. Moreover, PI3Kα/δ inhibition delivered strong single agent anti-tumor activity, which was associated with dynamic suppression of T-regs, improved CD8+ T-cell activation and memory in mouse syngeneic tumor models. Strikingly, AZD8835 promoted robust CD8+ T-cell activation dissociated from its effect on T-regs. This was associated with enhancing effector cell viability/function. Together these data reveal novel mechanisms by which PI3Kα/δ inhibitors interact with the immune system and validate the clinical compound AZD8835 as a novel immunoncology drug, independent of effects on tumor cells. These data support further clinical investigation of PI3K pathway inhibitors as immuno-oncology agents.
Collapse
Affiliation(s)
- Larissa S Carnevalli
- Bioscience, Oncology, IMED Biotech Unit AstraZeneca, Francis Crick Ave, Cambridge, CB2 0SL, UK.
| | - Charles Sinclair
- Bioscience, Oncology, IMED Biotech Unit AstraZeneca, Francis Crick Ave, Cambridge, CB2 0SL, UK
| | - Molly A Taylor
- Bioscience, Oncology, IMED Biotech Unit AstraZeneca, Francis Crick Ave, Cambridge, CB2 0SL, UK
| | | | - Sophie Langdon
- Bioscience, Oncology, IMED Biotech Unit AstraZeneca, Francis Crick Ave, Cambridge, CB2 0SL, UK.,Present Address: University of Birmingham, B15 2TT, Birmingham, UK
| | - Anna M L Coenen-Stass
- Bioscience, Oncology, IMED Biotech Unit AstraZeneca, Francis Crick Ave, Cambridge, CB2 0SL, UK
| | - Lorraine Mooney
- Bioscience, Oncology, IMED Biotech Unit AstraZeneca, Alderley Park, Alderley Edge, Macclesfield, SK10 4TG, UK.,Present Address: Alderley Park Limited, Preclinical Services, Alderley Park, Macclesfield, SK10 4TG, UK
| | - Adina Hughes
- Bioscience, Oncology, IMED Biotech Unit AstraZeneca, Francis Crick Ave, Cambridge, CB2 0SL, UK
| | - Laura Jarvis
- Bioscience, Oncology, IMED Biotech Unit AstraZeneca, Francis Crick Ave, Cambridge, CB2 0SL, UK
| | - Anna Staniszewska
- Bioscience, Oncology, IMED Biotech Unit AstraZeneca, Francis Crick Ave, Cambridge, CB2 0SL, UK
| | - Claire Crafter
- Bioscience, Oncology, IMED Biotech Unit AstraZeneca, Francis Crick Ave, Cambridge, CB2 0SL, UK
| | - Ben Sidders
- Bioscience, Oncology, IMED Biotech Unit AstraZeneca, Francis Crick Ave, Cambridge, CB2 0SL, UK
| | - Elizabeth Hardaker
- Bioscience, Oncology, IMED Biotech Unit AstraZeneca, Francis Crick Ave, Cambridge, CB2 0SL, UK
| | - Kevin Hudson
- Bioscience, Oncology, IMED Biotech Unit AstraZeneca, Alderley Park, Alderley Edge, Macclesfield, SK10 4TG, UK.,Present Address: 2theNth, Adelphi Group, Bollington, SK10 5JB, UK
| | - Simon T Barry
- Bioscience, Oncology, IMED Biotech Unit AstraZeneca, Francis Crick Ave, Cambridge, CB2 0SL, UK.
| |
Collapse
|
48
|
Pilipow K, Scamardella E, Puccio S, Gautam S, De Paoli F, Mazza EM, De Simone G, Polletti S, Buccilli M, Zanon V, Di Lucia P, Iannacone M, Gattinoni L, Lugli E. Antioxidant metabolism regulates CD8+ T memory stem cell formation and antitumor immunity. JCI Insight 2018; 3:122299. [PMID: 30232291 DOI: 10.1172/jci.insight.122299] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/07/2018] [Indexed: 12/17/2022] Open
Abstract
Adoptive T cell transfer (ACT) immunotherapy benefits from early differentiated stem cell memory T (Tscm) cells capable of persisting in the long term and generating potent antitumor effectors. Due to their paucity ex vivo, Tscm cells can be derived from naive precursors, but the molecular signals at the basis of Tscm cell generation are ill-defined. We found that less differentiated human circulating CD8+ T cells display substantial antioxidant capacity ex vivo compared with more differentiated central and effector memory T cells. Limiting ROS metabolism with antioxidants during naive T cell activation hindered terminal differentiation, while allowing expansion and generation of Tscm cells. N-acetylcysteine (NAC), the most effective molecule in this regard, induced transcriptional and metabolic programs characteristic of self-renewing memory T cells. Upon ACT, NAC-generated Tscm cells established long-term memory in vivo and exerted more potent antitumor immunity in a xenogeneic model when redirected with CD19-specific CAR, highlighting the translational relevance of NAC as a simple and inexpensive method to improve ACT.
Collapse
Affiliation(s)
- Karolina Pilipow
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Eloise Scamardella
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Simone Puccio
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Sanjivan Gautam
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Federica De Paoli
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Emilia Mc Mazza
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Gabriele De Simone
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | | | - Marta Buccilli
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Veronica Zanon
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Pietro Di Lucia
- Division of Immunology, Transplantation and Infectious Diseases and Experimental Imaging Center, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milan, Italy
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases and Experimental Imaging Center, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milan, Italy
| | - Luca Gattinoni
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Enrico Lugli
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Humanitas Flow Cytometry Core, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| |
Collapse
|
49
|
Ajina A, Maher J. Strategies to Address Chimeric Antigen Receptor Tonic Signaling. Mol Cancer Ther 2018; 17:1795-1815. [PMID: 30181329 PMCID: PMC6130819 DOI: 10.1158/1535-7163.mct-17-1097] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/19/2018] [Accepted: 06/18/2018] [Indexed: 12/18/2022]
Abstract
Adoptive cell transfer using chimeric antigen receptors (CAR) has emerged as one of the most promising new therapeutic modalities for patients with relapsed or refractory B-cell malignancies. Thus far, results in patients with advanced solid tumors have proven disappointing. Constitutive tonic signaling in the absence of ligand is an increasingly recognized complication when deploying these synthetic fusion receptors and can be a cause of poor antitumor efficacy, impaired survival, and reduced persistence in vivo In parallel, ligand-dependent tonic signaling can mediate toxicity and promote T-cell anergy, exhaustion, and activation-induced cell death. Here, we review the mechanisms underpinning CAR tonic signaling and highlight the wide variety of effects that can emerge after making subtle structural changes or altering the methodology of CAR transduction. We highlight strategies to prevent unconstrained tonic signaling and address its deleterious consequences. We also frame this phenomenon in the context of endogenous TCR tonic signaling, which has been shown to regulate peripheral tolerance, facilitate the targeting of foreign antigens, and suggest opportunities to coopt ligand-dependent CAR tonic signaling to facilitate in vivo persistence and efficacy. Mol Cancer Ther; 17(9); 1795-815. ©2018 AACR.
Collapse
MESH Headings
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Humans
- Immunotherapy, Adoptive/methods
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/metabolism
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Adam Ajina
- CAR Mechanics Group, King's College London, London, United Kingdom.
- School of Cancer and Pharmaceutical Studies, Guy's Hospital, London, United Kingdom
| | - John Maher
- CAR Mechanics Group, King's College London, London, United Kingdom
- School of Cancer and Pharmaceutical Studies, Guy's Hospital, London, United Kingdom
- Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, United Kingdom
- Department of Immunology, Eastbourne Hospital, East Sussex, United Kingdom
| |
Collapse
|
50
|
Ghassemi S, Nunez-Cruz S, O'Connor RS, Fraietta JA, Patel PR, Scholler J, Barrett DM, Lundh SM, Davis MM, Bedoya F, Zhang C, Leferovich J, Lacey SF, Levine BL, Grupp SA, June CH, Melenhorst JJ, Milone MC. Reducing Ex Vivo Culture Improves the Antileukemic Activity of Chimeric Antigen Receptor (CAR) T Cells. Cancer Immunol Res 2018; 6:1100-1109. [PMID: 30030295 PMCID: PMC8274631 DOI: 10.1158/2326-6066.cir-17-0405] [Citation(s) in RCA: 183] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/22/2017] [Accepted: 07/16/2018] [Indexed: 12/24/2022]
Abstract
The success of chimeric antigen receptor (CAR)-mediated immunotherapy in acute lymphoblastic leukemia (ALL) highlights the potential of T-cell therapies with directed cytotoxicity against specific tumor antigens. The efficacy of CAR T-cell therapy depends on the engraftment and persistence of T cells following adoptive transfer. Most protocols for T-cell engineering routinely expand T cells ex vivo for 9 to 14 days. Because the potential for engraftment and persistence is related to the state of T-cell differentiation, we hypothesized that reducing the duration of ex vivo culture would limit differentiation and enhance the efficacy of CAR T-cell therapy. We demonstrated that T cells with a CAR-targeting CD19 (CART19) exhibited less differentiation and enhanced effector function in vitro when harvested from cultures at earlier (day 3 or 5) compared with later (day 9) timepoints. We then compared the therapeutic potential of early versus late harvested CART19 in a murine xenograft model of ALL and showed that the antileukemic activity inversely correlated with ex vivo culture time: day 3 harvested cells showed robust tumor control despite using a 6-fold lower dose of CART19, whereas day 9 cells failed to control leukemia at limited cell doses. We also demonstrated the feasibility of an abbreviated culture in a large-scale current good manufacturing practice-compliant process. Limiting the interval between T-cell isolation and CAR treatment is critical for patients with rapidly progressing disease. Generating CAR T cells in less time also improves potency, which is central to the effectiveness of these therapies. Cancer Immunol Res; 6(9); 1100-9. ©2018 AACR.
Collapse
Affiliation(s)
- Saba Ghassemi
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Selene Nunez-Cruz
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Roddy S O'Connor
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joseph A Fraietta
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Prachi R Patel
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John Scholler
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David M Barrett
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Stefan M Lundh
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Megan M Davis
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Felipe Bedoya
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Changfeng Zhang
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John Leferovich
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Simon F Lacey
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bruce L Levine
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stephan A Grupp
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, Pennsylvania
| | - J Joseph Melenhorst
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael C Milone
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|