1
|
Qi S, Li J, Gu X, Zhang Y, Zhou W, Wang F, Wang W. Impacts of ageing on the efficacy of CAR-T cell therapy. Ageing Res Rev 2025; 107:102715. [PMID: 40058461 DOI: 10.1016/j.arr.2025.102715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/19/2025] [Accepted: 03/02/2025] [Indexed: 03/17/2025]
Abstract
Chimeric antigen receptor T cells recognizing CD19 (19CAR-T) cell therapy has achieved robust clinical efficacy when treating some hematological malignancies, but which patient subgroups benefit mostly remains elusive. Here we summarized the data of 541 patients from 30 clinical trials who underwent 19 CAR-T therapy and analyzed the different clinical responses between young (<44 years), middle-aged (45-59 years) and elderly (>60 years) patients and found that the young patients showed a higher level of complete response (CR) rate. Therefore, we then summarize the advances of studies focusing on the effects of age on anti-tumor efficacy of CAR-T therapy and analyze the reasons for the low CR rate after CAR-T cell therapy in elderly patients with tumors, aiming to provide hints for oncologists to select the most suitable candidate for this cancer immunotherapy.
Collapse
Affiliation(s)
- Shimao Qi
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Jiaqian Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Xinyu Gu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Yalan Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Weilin Zhou
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Fengling Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Wei Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
2
|
Pinto E, Lione L, Compagnone M, Paccagnella M, Salvatori E, Greco M, Frezza V, Marra E, Aurisicchio L, Roscilli G, Conforti A. From ex vivo to in vivo chimeric antigen T cells manufacturing: new horizons for CAR T-cell based therapy. J Transl Med 2025; 23:10. [PMID: 39755643 PMCID: PMC11700462 DOI: 10.1186/s12967-024-06052-3] [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: 09/19/2024] [Accepted: 12/24/2024] [Indexed: 01/06/2025] Open
Abstract
In the past decades, Chimeric Antigen Receptor (CAR)-T cell therapy has achieved remarkable success, leading to the approval of six therapeutic products for haematological malignancies. Recently, the therapeutic potential of this therapy has also been demonstrated in non-tumoral diseases. Currently, the manufacturing process to produce clinical-grade CAR-T cells is complex, time-consuming, and highly expensive. It involves multiple steps, including the collection of T cells from patients or healthy donors, in vitro engineering and expansion, and finally reinfusion into patients. Therefore, despite the impressive clinical outcomes, ex vivo manufacturing process makes CAR-T cells out of reach for many cancer patients. Direct in vivo engineering of T cells could be a more rapid solution able to circumvent both the complexity and the costs associated with ex vivo manufactured CAR-T cells. This novel approach allows to completely eliminate ex vivo cell manipulation and expansion while producing therapeutic cell populations directly in vivo. To date, several studies have demonstrated the feasibility of in vivo T cell reprogramming, by employing injectable viral- or nanocarrier-based delivery platforms in tumour animal models. Additionally, in vivo production of CAR-T cells might reduce the incidence, or at least the severity, of systemic toxicities frequently occurring with ex vivo produced CAR-T cells, such as cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. In this review, we highlight the challenges associated with the current ex vivo manufacturing protocols and review the latest progresses in the emerging field of in vivo CAR-T therapy, by comparing the various platforms so far investigated. Moreover, we offer an overview of the advantages deriving from in vivo reprogramming of other immune cell types, such as Natural Killer and macrophages, with CAR constructs.
Collapse
Affiliation(s)
- E Pinto
- Evvivax Biotech, Via Castel Romano 100, 00128, Rome, Italy
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - L Lione
- Takis Biotech, Via Castel Romano 100, 00128, Rome, Italy
| | - M Compagnone
- Evvivax Biotech, Via Castel Romano 100, 00128, Rome, Italy
| | - M Paccagnella
- Takis Biotech, Via Castel Romano 100, 00128, Rome, Italy
| | - E Salvatori
- Takis Biotech, Via Castel Romano 100, 00128, Rome, Italy
| | - M Greco
- Takis Biotech, Via Castel Romano 100, 00128, Rome, Italy
| | - V Frezza
- Takis Biotech, Via Castel Romano 100, 00128, Rome, Italy
| | - E Marra
- Takis Biotech, Via Castel Romano 100, 00128, Rome, Italy
| | - L Aurisicchio
- Evvivax Biotech, Via Castel Romano 100, 00128, Rome, Italy
- Takis Biotech, Via Castel Romano 100, 00128, Rome, Italy
| | - G Roscilli
- Takis Biotech, Via Castel Romano 100, 00128, Rome, Italy
| | - A Conforti
- Evvivax Biotech, Via Castel Romano 100, 00128, Rome, Italy.
- Takis Biotech, Via Castel Romano 100, 00128, Rome, Italy.
| |
Collapse
|
3
|
Hu M, Deng F, Song X, Zhao H, Yan F. The crosstalk between immune cells and tumor pyroptosis: advancing cancer immunotherapy strategies. J Exp Clin Cancer Res 2024; 43:190. [PMID: 38987821 PMCID: PMC11234789 DOI: 10.1186/s13046-024-03115-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024] Open
Abstract
Pyroptosis is a cell death process characterized by cell swelling until membrane rupture and release of intracellular contents. As an effective tumor treatment strategy, inducing tumor cell pyroptosis has received widespread attention. In this process, the immune components within the tumor microenvironment play a key regulatory role. By regulating and altering the functions of immune cells such as cytotoxic T lymphocytes, natural killer cells, tumor-associated macrophages, and neutrophils, tumor cell pyroptosis can be induced. This article provides a comprehensive review of the molecular mechanisms of cell pyroptosis, the impact of the tumor immune microenvironment on tumor cell pyroptosis, and its mechanisms. It aims to gain an in-depth understanding of the communication between the tumor immune microenvironment and tumor cells, and to provide theoretical support for the development of new tumor immunotherapies.
Collapse
Affiliation(s)
- Mengyuan Hu
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Chenggong District, 1168 Chunrong West Road, Yunhua Street, Kunming, 650500, Yunnan, China
| | - Fengying Deng
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Chenggong District, 1168 Chunrong West Road, Yunhua Street, Kunming, 650500, Yunnan, China
| | - Xinlei Song
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Chenggong District, 1168 Chunrong West Road, Yunhua Street, Kunming, 650500, Yunnan, China
| | - Hongkun Zhao
- Key Laboratory of Yunnan Province, Yunnan Eye Institute, Affiliated Hospital of Yunnan University, Yunnan University, 176 Qingnian Road, Wuhua District, Kunming, 650031, Yunnan, China.
| | - Fei Yan
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Chenggong District, 1168 Chunrong West Road, Yunhua Street, Kunming, 650500, Yunnan, China.
| |
Collapse
|
4
|
Khoshandam M, Soltaninejad H, Hamidieh AA, Hosseinkhani S. CRISPR, CAR-T, and NK: Current applications and future perspectives. Genes Dis 2024; 11:101121. [PMID: 38545126 PMCID: PMC10966184 DOI: 10.1016/j.gendis.2023.101121] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 08/16/2023] [Indexed: 11/11/2024] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy represents a breakthrough in personalized cancer treatments. In this regard, synthetic receptors comprised of antigen recognition domains, signaling, and stimulatory domains are used to reprogram T-cells to target tum or cells and destroy them. Despite the success of this approach in refractory B-cell malignancies, the optimal potency of CAR T-cell therapy for many other cancers, particularly solid tumors, has not been validated. Natural killer cells are powerful cytotoxic lymphocytes specialized in recognizing and dispensing the tumor cells in coordination with other anti-tumor immunity cells. Based on these studies, many investigations are focused on the accurate designing of CAR T-cells with clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system or other novel gene editing tools that can induce hereditary changes with or without the presence of a double-stranded break into the genome. These methodologies can be specifically focused on negative controllers of T-cells, induce modifications to a particular gene, and produce reproducible, safe, and powerful allogeneic CAR T-cells for on-demand cancer immunotherapy. The improvement of the CRISPR/Cas9 innovation offers an adaptable and proficient gene-editing capability in activating different pathways to help natural killer cells interact with novel CARs to particularly target tumor cells. Novel achievements and future challenges of combining next-generation CRISPR-Cas9 gene editing tools to optimize CAR T-cell and natural killer cell treatment for future clinical trials toward the foundation of modern cancer treatments have been assessed in this review.
Collapse
Affiliation(s)
- Mohadeseh Khoshandam
- Department of Reproductive Biology, Academic Center for Education, Culture, and Research (ACECR), Qom branch 3716986466, Iran
- National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran 14965/161, Iran
| | - Hossein Soltaninejad
- Department of stem cells technology and Tissue Regeneration, Faculty of Interdisciplinary Science and Technologies, Tarbiat Modares University, Tehran 15614, Iran
- Pediatric Cell Therapy and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran 1417935840, Iran
| | - Amir Ali Hamidieh
- Pediatric Cell Therapy and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran 1417935840, Iran
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 15614, Iran
| |
Collapse
|
5
|
Mardi A, Shirokova AV, Mohammed RN, Keshavarz A, Zekiy AO, Thangavelu L, Mohamad TAM, Marofi F, Shomali N, Zamani A, Akbari M. Biological causes of immunogenic cancer cell death (ICD) and anti-tumor therapy; Combination of Oncolytic virus-based immunotherapy and CAR T-cell therapy for ICD induction. Cancer Cell Int 2022; 22:168. [PMID: 35488303 PMCID: PMC9052538 DOI: 10.1186/s12935-022-02585-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 04/11/2022] [Indexed: 12/22/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a promising and rapidly expanding therapeutic option for a wide range of human malignancies. Despite the ongoing progress of CAR T-cell therapy in hematologic malignancies, the application of this therapeutic strategy in solid tumors has encountered several challenges due to antigen heterogeneity, suboptimal CAR T-cell trafficking, and the immunosuppressive features of the tumor microenvironment (TME). Oncolytic virotherapy is a novel cancer therapy that employs competent or genetically modified oncolytic viruses (OVs) to preferentially proliferate in tumor cells. OVs in combination with CAR T-cells are promising candidates for overcoming the current drawbacks of CAR T-cell application in tumors through triggering immunogenic cell death (ICD) in cancer cells. ICD is a type of cellular death in which danger-associated molecular patterns (DAMPs) and tumor-specific antigens are released, leading to the stimulation of potent anti-cancer immunity. In the present review, we discuss the biological causes of ICD, different types of ICD, and the synergistic combination of OVs and CAR T-cells to reach potent tumor-specific immunity.
Collapse
Affiliation(s)
- Amirhossein Mardi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Anastasia V Shirokova
- Department of Prosthetic Dentistry, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Rebar N Mohammed
- Medical Laboratory Analysis Department, College of Health Science, Cihan University of Sulaimaniya, Suleimanyah, Kurdistan region, Iraq.,College of. Veterinary Medicine, University of Sulaimani, Suleimanyah, Iraq
| | - Ali Keshavarz
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Angelina O Zekiy
- Department of Prosthetic Dentistry, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, India
| | - Talar Ahmad Merza Mohamad
- Department of Pharmacology and Toxicology, Clinical Pharmacy, Hawler Medical University, College of Pharmacy, Kurdistan Region-Erbil, Iraq
| | - Faroogh Marofi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zamani
- Shiraz Transplant Center, Abu Ali Sina Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Morteza Akbari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
Stock S, Kluever AK, Endres S, Kobold S. Enhanced Chimeric Antigen Receptor T Cell Therapy through Co-Application of Synergistic Combination Partners. Biomedicines 2022; 10:biomedicines10020307. [PMID: 35203517 PMCID: PMC8869718 DOI: 10.3390/biomedicines10020307] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 11/16/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has achieved remarkable response rates and revolutionized the treatment of patients suffering from defined hematological malignancies. However, many patients still do not respond to this therapy or relapse after an initial remission, underscoring the need for improved efficacy. Insufficient in vivo activity, persistence, trafficking, and tumor infiltration of CAR T cells, as well as antigen escape and treatment-associated adverse events, limit the therapeutic success. Multiple strategies and approaches have been investigated to further improve CAR T cell therapy. Besides genetic modification of the CAR itself, the combination with other treatment modalities has the potential to improve this approach. In particular, combining CAR T cells with clinically approved compounds such as monoclonal antibodies and small molecule inhibitors might be a promising strategy. Combination partners could already be applied during the production process to influence the cellular composition and immunophenotype of the final CAR T cell product. Alternatively, simultaneous administration of clinically approved compounds with CAR T cells would be another feasible avenue. In this review, we will discuss current strategies to combine CAR T cells with compounds to overcome recent limitations and further enhance this promising cancer therapy, potentially broadening its application beyond hematology.
Collapse
Affiliation(s)
- Sophia Stock
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, 80337 Munich, Germany; (A.-K.K.); (S.E.)
- Department of Medicine III, University Hospital, Ludwig Maximilian University (LMU) of Munich, 81337 Munich, Germany
- Correspondence: (S.S.); (S.K.)
| | - Anna-Kristina Kluever
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, 80337 Munich, Germany; (A.-K.K.); (S.E.)
| | - Stefan Endres
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, 80337 Munich, Germany; (A.-K.K.); (S.E.)
- German Center for Translational Cancer Research (DKTK), Partner Site Munich, 80336 Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), 85764 Neuherberg, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, 80337 Munich, Germany; (A.-K.K.); (S.E.)
- German Center for Translational Cancer Research (DKTK), Partner Site Munich, 80336 Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), 85764 Neuherberg, Germany
- Correspondence: (S.S.); (S.K.)
| |
Collapse
|
7
|
Skurikhin E, Pershina O, Zhukova M, Widera D, Ermakova N, Pan E, Pakhomova A, Morozov S, Kubatiev A, Dygai A. Potential of Stem Cells and CART as a Potential Polytherapy for Small Cell Lung Cancer. Front Cell Dev Biol 2021; 9:778020. [PMID: 34926461 PMCID: PMC8678572 DOI: 10.3389/fcell.2021.778020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/18/2021] [Indexed: 12/15/2022] Open
Abstract
Despite the increasing urgency of the problem of treating small cell lung cancer (SCLC), information on the causes of its development is fragmentary. There is no complete understanding of the features of antitumor immunity and the role of the microenvironment in the development of SCLC resistance. This impedes the development of new methods for the diagnosis and treatment of SCLC. Lung cancer and chronic obstructive pulmonary disease (COPD) have common pathogenetic factors. COPD is a risk factor for lung cancer including SCLC. Therefore, the search for effective approaches to prevention, diagnosis, and treatment of SCLC in patients with COPD is an urgent task. This review provides information on the etiology and pathogenesis of SCLC, analyses the effectiveness of current treatment options, and critically evaluates the potential of chimeric antigen receptor T cells therapy (CART therapy) in SCLC. Moreover, we discuss potential links between lung cancer and COPD and the role of endothelium in the development of COPD. Finally, we propose a new approach for increasing the efficacy of CART therapy in SCLC.
Collapse
Affiliation(s)
- Evgenii Skurikhin
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Olga Pershina
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Mariia Zhukova
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Darius Widera
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, University of Reading, Reading, United Kingdom
| | - Natalia Ermakova
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Edgar Pan
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Angelina Pakhomova
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Sergey Morozov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Aslan Kubatiev
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Alexander Dygai
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| |
Collapse
|
8
|
Wang S, Sellner L, Wang L, Sauer T, Neuber B, Gong W, Stock S, Ni M, Yao H, Kleist C, Schmitt A, Müller-Tidow C, Schmitt M, Schubert ML. Combining selective inhibitors of nuclear export (SINEs) with chimeric antigen receptor (CAR) T cells for CD19‑positive malignancies. Oncol Rep 2021; 46:170. [PMID: 34165175 PMCID: PMC8250584 DOI: 10.3892/or.2021.8121] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 04/08/2021] [Indexed: 11/06/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells directed against CD19 (CD19.CAR T cells) have yielded impressive clinical responses in the treatment of patients with lymphoid malignancies. However, resistance and/or relapse can limit treatment outcome. Risk of tumor escape can be reduced by combining treatment strategies. Selective inhibitors of nuclear export (SINEs) directed against nuclear exportin‑1 (XPO1) have demonstrated anti‑tumor efficacy in several hematological malignancies. The aim of the present study was to evaluate the combination of CAR T cells with the SINE compounds eltanexor and selinexor. As expected, eltanexor and selinexor were toxic to CD19‑positive malignant cells and the sensitivity of cells towards SINEs correlated with the levels of XPO1‑expression in ALL cell lines. When SINEs and CAR T cells were simultaneously combined, SINEs exerted toxicity towards CAR T cells and impaired their function affecting cytotoxicity and cytokine release ability. Flow cytometry and western blot analysis revealed that eltanexor decreased the cytoplasmic concentration of the transcription factor phosphorylated‑STAT3 in CAR T cells. Due to CAR T‑cell toxicity, sequential use of SINEs and CAR T cells was evaluated: Cytotoxicity of CAR T cells increased significantly when target cells were pre‑treated with the SINE compound eltanexor. In addition, exhaustion of CAR T cells decreased when target cells were pre‑treated with eltanexor. In summary, whereas the concomitant use of SINEs and CAR T cells does not seem advisable, sequential use of SINEs and CAR T cells might improve the anti‑tumor efficacy of CAR T cells.
Collapse
Affiliation(s)
- Sanmei Wang
- Department of Internal Medicine V, Heidelberg University Hospital, D-69120 Heidelberg, Germany
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Leopold Sellner
- Department of Internal Medicine V, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| | - Lei Wang
- Department of Internal Medicine V, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| | - Tim Sauer
- Department of Internal Medicine V, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| | - Brigitte Neuber
- Department of Internal Medicine V, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| | - Wenjie Gong
- Department of Internal Medicine V, Heidelberg University Hospital, D-69120 Heidelberg, Germany
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Sophia Stock
- Department of Internal Medicine V, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| | - Ming Ni
- Department of Internal Medicine V, Heidelberg University Hospital, D-69120 Heidelberg, Germany
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Hao Yao
- Department of Internal Medicine V, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| | - Christian Kleist
- Department of Nuclear Medicine, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, D-69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT)/German Cancer Consortium (DKTK), D-69120 Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, D-69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT)/German Cancer Consortium (DKTK), D-69120 Heidelberg, Germany
| | - Maria-Luisa Schubert
- Department of Internal Medicine V, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| |
Collapse
|
9
|
Lu W, Wei Y, Cao Y, Xiao X, Li Q, Lyu H, Jiang Y, Zhang H, Li X, Jiang Y, Meng J, Yuan T, Zhu H, He X, Jin X, Sun R, Sui T, Liu K, Zhao M. CD19 CAR-T cell treatment conferred sustained remission in B-ALL patients with minimal residual disease. Cancer Immunol Immunother 2021; 70:3501-3511. [PMID: 33899130 PMCID: PMC8571234 DOI: 10.1007/s00262-021-02941-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 04/09/2021] [Indexed: 01/23/2023]
Abstract
The persistence or recurrence of minimal residual disease (MRD) after chemotherapy predicts relapse of B-cell acute lymphoblastic leukemia (B-ALL). CD19-directed chimeric antigen receptor T (CD19 CAR-T) cells have shown promising responses in B-ALL. However, their role in chemotherapy-refractory MRD-positive B-ALL remains unclear. Here we aimed to assess the effectiveness and safety of CD19 CAR-T cells in MRD-positive B-ALL patients. From January 2018, a total of 14 MRD-positive B-ALL patients received one or more infusions of autogenous CD19 CAR-T cells. Among them, 12 patients achieved MRD-negative remission after one cycle of CAR-T infusion. At a median follow-up time of 647 days (range 172–945 days), the 2-year event-free survival rate in MRD-positive patients was 61.2% ± 14.0% and the 2-year overall survival was 78.6 ± 11.0%, which were significantly higher than patients with active disease (blasts ≥ 5% or with extramedullary disease). Moreover, patients with MRD had a lower grade of cytokine release syndrome (CRS) than patients with active disease. However, the peak expansion of CAR-T cells in MRD positive patients showed no statistical difference compared to patients with active disease. Five patients received two or more CAR-T cell infusions and these patients showed a decreased peak expansion of CAR-T cell in subsequent infusions. In conclusion, pre-emptive CD19 CAR-T cell treatment is an effective and safe approach and may confer sustained remission in B-ALL patients with chemotherapy-refractory MRD. The trials were registered at www.chictr.org.cn as ChiCTR-ONN-16009862 (November 14, 2016) and ChiCTR1800015164 (March 11, 2018).
Collapse
Affiliation(s)
- Wenyi Lu
- Department of Hematology, Tianjin First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China.,Nankai University Affiliated First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China
| | - Yunxiong Wei
- Department of Hematology, Tianjin First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China
| | - Yaqing Cao
- Department of Hematology, Tianjin First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China
| | - Xia Xiao
- Department of Hematology, Tianjin First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China.,Nankai University Affiliated First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China
| | - Qing Li
- Department of Hematology, Tianjin First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China.,Nankai University Affiliated First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China
| | - Hairong Lyu
- Department of Hematology, Tianjin First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China.,Nankai University Affiliated First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China
| | - Yili Jiang
- Department of Hematology, Tianjin First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China.,Nankai University Affiliated First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China
| | - Huan Zhang
- Department of Hematology, Tianjin First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China.,Nankai University Affiliated First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China
| | - Xin Li
- Department of Hematology, Tianjin First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China.,Nankai University Affiliated First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China
| | - Yanyu Jiang
- Department of Hematology, Tianjin First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China.,Nankai University Affiliated First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China
| | - Juanxia Meng
- Department of Hematology, Tianjin First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China.,Nankai University Affiliated First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China
| | - Ting Yuan
- Department of Hematology, Tianjin First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China.,Nankai University Affiliated First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China
| | - Haibo Zhu
- Department of Hematology, Tianjin First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China.,Nankai University Affiliated First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China
| | - Xiaoyuan He
- Nankai University Affiliated First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China
| | - Xin Jin
- School of Medicine, Nankai University, No.94 Weijin Road, Tianjin, 300071, People's Republic of China
| | - Rui Sun
- Department of Hematology, Tianjin First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China
| | - Tao Sui
- Department of Hematology, Tianjin First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China.,Nankai University Affiliated First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China
| | - Kaiqi Liu
- Leukemia Center, Institute of Hematology and Blood Diseases Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 288 Nanjing Road, Tianjin, 300060, People's Republic of China.
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China. .,Nankai University Affiliated First Central Hospital, No. 24 Fu Kang Road, Tianjin, 300192, People's Republic of China.
| |
Collapse
|
10
|
Luo Y, Song G, Liang S, Li F, Liu K. Research advances in chimeric antigen receptor-modified T-cell therapy (Review). Exp Ther Med 2021; 21:484. [PMID: 33790993 PMCID: PMC8005741 DOI: 10.3892/etm.2021.9915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
Chimeric antigen receptor (CAR)-modified T-cells are T-cells that have been genetically engineered to express CAR molecules to target specific surface antigens on tumor cells. CAR T-cell therapy, a novel cancer immunotherapy, has been attracting increasing attention, since it exhibited notable efficacy in the treatment of hematological tumors in clinical trials. However, for this type of therapy, challenges must be overcome in the treatment of solid tumors. Furthermore, certain side effects associated with CAR T-cell therapy, including cytokine release syndrome, immune effector cell-related neurotoxicity syndrome, tumor lysis syndrome and on-target off-tumor toxicity, must be taken into consideration. The present study provides a systematic review of the principle, clinical application, current challenges, possible solutions and future perspectives for CAR T-cell therapy.
Collapse
Affiliation(s)
- Yuxi Luo
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China.,The First Clinic of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Guiqin Song
- Department of Biology, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Shichu Liang
- The First Clinic of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Feifei Li
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
11
|
Current Challenges in Providing Good Leukapheresis Products for Manufacturing of CAR-T Cells for Patients with Relapsed/Refractory NHL or ALL. Cells 2020; 9:cells9051225. [PMID: 32429189 PMCID: PMC7290830 DOI: 10.3390/cells9051225] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/09/2020] [Accepted: 05/13/2020] [Indexed: 12/15/2022] Open
Abstract
Background: T lymphocyte collection through leukapheresis is an essential step for chimeric antigen receptor T (CAR-T) cell therapy. Timing of apheresis is challenging in heavily pretreated patients who suffer from rapid progressive disease and receive T cell impairing medication. Methods: A total of 75 unstimulated leukaphereses were analyzed including 45 aphereses in patients and 30 in healthy donors. Thereof, 41 adult patients with Non-Hodgkin’s lymphoma (85%) or acute lymphoblastic leukemia (15%) underwent leukapheresis for CAR-T cell production. Results: Sufficient lymphocytes were harvested from all patients even from those with low peripheral lymphocyte counts of 0.18/nL. Only four patients required a second leukapheresis session. Leukapheresis products contained a median of 98 × 108 (9 - 341 × 108) total nucleated cells (TNC) with 38 × 108 (4 - 232 × 108) CD3+ T cells. Leukapheresis products from healthy donors as well as from patients in complete remission were characterized by high TNC and CD3+ T lymphocyte counts. CAR-T cell products could be manufactured for all but one patient. Conclusions: Sufficient yield of lymphocytes for CAR-T cell production is feasible also for patients with low peripheral blood counts. Up to 12–15 L blood volume should be processed in patients with absolute lymphocyte counts ≤ 1.0/nL.
Collapse
|
12
|
Dreger P, Fenske TS, Montoto S, Pasquini MC, Sureda A, Hamadani M. Cellular Immunotherapy for Refractory Diffuse Large B Cell Lymphoma in the Chimeric Antigen Receptor-Engineered T Cell Era: Still a Role for Allogeneic Transplantation? Biol Blood Marrow Transplant 2020; 26:e77-e85. [PMID: 31917272 PMCID: PMC7207150 DOI: 10.1016/j.bbmt.2019.12.771] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/26/2019] [Accepted: 12/30/2019] [Indexed: 12/29/2022]
Abstract
Chimeric antigen receptor-engineered T (CART) cells are a promising new treatment option for patients with multiply relapsed and refractory (R/R) diffuse large B cell lymphoma (DLBCL). Because of the favorable outcome data reported for CART cells, uncertainty is emerging if there is still a role for allogeneic hematopoietic cell transplantation (allo-HCT) in the treatment of R/R DLBCL. This article provides an overview of available evidence and theoretical considerations to put these 2 types of cellular immunotherapy (CI) into perspective. Altogether, current data suggest that CART cells are preferred now over transplantation as first-choice CI in many clinical situations. However, the majority of patients will fail CART therapy, resulting in an unmet medical need where allo-HCT could be beneficial. In contrast, employing allo-HCT instead of CART cells as first CI should be presently restricted to situations where CART cell therapy is deemed not feasible or useful, such as patients with refractory cytopenia or incipient myelodysplastic syndrome. However, allo-HCT remains a standard treatment option as first CI for patients with chemosensitive R/R DLBCL when CARTs are not available or transplantation is preferred by the patient. Continuous collection and analysis of CI outcome data by professional registries appear to be of key importance for developing rational strategies of CI allocation and sequencing.
Collapse
Affiliation(s)
- Peter Dreger
- Department Medicine V, University of Heidelberg, Heidelberg, Germany.
| | - Timothy S Fenske
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Silvia Montoto
- Department of Haemato-oncology, St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Marcelo C Pasquini
- CIBMTR, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Anna Sureda
- Hematology Department, Institut Català d'Oncologia-Hospitalet, Barcelona, Spain
| | - Mehdi Hamadani
- CIBMTR, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
13
|
Fleischer LC, Spencer HT, Raikar SS. Targeting T cell malignancies using CAR-based immunotherapy: challenges and potential solutions. J Hematol Oncol 2019; 12:141. [PMID: 31884955 PMCID: PMC6936092 DOI: 10.1186/s13045-019-0801-y] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 10/09/2019] [Indexed: 12/23/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has been successful in treating B cell malignancies in clinical trials; however, fewer studies have evaluated CAR T cell therapy for the treatment of T cell malignancies. There are many challenges in translating this therapy for T cell disease, including fratricide, T cell aplasia, and product contamination. To the best of our knowledge, no tumor-specific antigen has been identified with universal expression on cancerous T cells, hindering CAR T cell therapy for these malignancies. Numerous approaches have been assessed to address each of these challenges, such as (i) disrupting target antigen expression on CAR-modified T cells, (ii) targeting antigens with limited expression on T cells, and (iii) using third party donor cells that are either non-alloreactive or have been genome edited at the T cell receptor α constant (TRAC) locus. In this review, we discuss CAR approaches that have been explored both in preclinical and clinical studies targeting T cell antigens, as well as examine other potential strategies that can be used to successfully translate this therapy for T cell disease.
Collapse
Affiliation(s)
- Lauren C Fleischer
- Molecular and Systems Pharmacology Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University School of Medicine, Atlanta, GA, USA
- Cell and Gene Therapy Program, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University School of Medicine, Atlanta, GA, USA
| | - H Trent Spencer
- Molecular and Systems Pharmacology Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University School of Medicine, Atlanta, GA, USA
- Cell and Gene Therapy Program, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University School of Medicine, Atlanta, GA, USA
| | - Sunil S Raikar
- Cell and Gene Therapy Program, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
14
|
Stock S, Schmitt M, Sellner L. Optimizing Manufacturing Protocols of Chimeric Antigen Receptor T Cells for Improved Anticancer Immunotherapy. Int J Mol Sci 2019; 20:ijms20246223. [PMID: 31835562 PMCID: PMC6940894 DOI: 10.3390/ijms20246223] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/07/2019] [Accepted: 12/08/2019] [Indexed: 01/08/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy can achieve outstanding response rates in heavily pretreated patients with hematological malignancies. However, relapses occur and they limit the efficacy of this promising treatment approach. The cellular composition and immunophenotype of the administered CART cells play a crucial role for therapeutic success. Less differentiated CART cells are associated with improved expansion, long-term in vivo persistence, and prolonged anti-tumor control. Furthermore, the ratio between CD4+ and CD8+ T cells has an effect on the anti-tumor activity of CART cells. The composition of the final cell product is not only influenced by the CART cell construct, but also by the culturing conditions during ex vivo T cell expansion. This includes different T cell activation strategies, cytokine supplementation, and specific pathway inhibition for the differentiation blockade. The optimal production process is not yet defined. In this review, we will discuss the use of different CART cell production strategies and the molecular background for the generation of improved CART cells in detail.
Collapse
Affiliation(s)
- Sophia Stock
- Department of Internal Medicine V, Heidelberg University Hospital, 69120 Heidelberg, Germany; (S.S.); (M.S.)
| | - Michael Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, 69120 Heidelberg, Germany; (S.S.); (M.S.)
- National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Leopold Sellner
- Department of Internal Medicine V, Heidelberg University Hospital, 69120 Heidelberg, Germany; (S.S.); (M.S.)
- National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- Oncology Business Unit—Medical Affairs, Takeda Pharma Vertrieb GmbH & Co. KG, 10117 Berlin, Germany
- Correspondence:
| |
Collapse
|
15
|
Hao L, Li T, Chang LJ, Chen X. Adoptive Immunotherapy for B-cell Malignancies Using CD19- Targeted Chimeric Antigen Receptor T-Cells: A Systematic Review of Efficacy and Safety. Curr Med Chem 2019; 26:3068-3079. [PMID: 28762313 DOI: 10.2174/0929867324666170801101842] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 06/15/2017] [Accepted: 07/25/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Adoptive infusion of chimeric antigen receptor transduced T- cells (CAR-T) is a powerful tool of immunotherapy for hematological malignancies, as evidenced by recently published and unpublished clinical results. OBJECTIVE In this report, we performed a meta-analysis to evaluate the efficacy and side effects of CAR-T on refractory and/or relapsed B-cell malignancies, including leukemia and lymphoma. METHODS Clinical studies investigating efficacy and safety of CAR-T in acute and chronic lymphocytic leukemia and lymphoma were identified by searching PubMed and EMBASE. Outcomes of efficacy subjected to analysis were the rates of complete remission (CR) and partial remission (PR). The safety parameters were the prevalence of adverse effects including fever, hypotension, and acute renal failure. Meta analyses were performed using R software. Weighted hazard ratio (HR) with 95% confidence intervals was calculated for each outcome. Fixed or random-effects models were employed depending on the heterogeneity across the included studies. RESULTS Nineteen published clinical studies with a total of 391 patients were included for the meta-analysis. The pooled rate of complete remission was 55% (95% CI 41%-69%); the pooled rate of partial remission was 25% (95% CI: 19%-33%). The prevalence of fever was 62% (95% CI: 41%-79%), the hypotension was 22% (95% CI: 15%-31%), and the acute renal failure was 24% (95% CI: 16%-34%). All adverse effects were manageable and no death was reported due to toxicity. CONCLUSION CD19-targeted CAR-T is an effective modality in treating refractory B-cell malignancies including leukemia and lymphoma. However, there is still a need to develop strategies to improve the safety in its clinical use.
Collapse
Affiliation(s)
- Lu Hao
- Shenzhen Geno-Immune Medical Institute, Shenzhen 518057, China.,Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China
| | - Tongtong Li
- Clinical Medicine Program, Nanchang University Medical College, Nanchang 330006, China.,Department of Obstetrics and Gynecology, Anfu People's Hospital, Jiangxi Province 343200, China
| | - Lung-Ji Chang
- Shenzhen Geno-Immune Medical Institute, Shenzhen 518057, China.,Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32610, United States
| | - Xiaochuan Chen
- Shenzhen Geno-Immune Medical Institute, Shenzhen 518057, China.,Department of Oriental Medicine, New York College of Health Professions, New York, NY 10016, United States
| |
Collapse
|
16
|
Marei HE, Althani A, Caceci T, Arriga R, Sconocchia T, Ottaviani A, Lanzilli G, Roselli M, Caratelli S, Cenciarelli C, Sconocchia G. Recent perspective on CAR and Fcγ-CR T cell immunotherapy for cancers: Preclinical evidence versus clinical outcomes. Biochem Pharmacol 2019; 166:335-346. [PMID: 31176617 DOI: 10.1016/j.bcp.2019.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 06/03/2019] [Indexed: 12/16/2022]
Abstract
The chimeric antigen receptor T cell (CAR-T cell) immunotherapy currently represents a hot research trend and it is expected to revolutionize the field of cancer therapy. Promising outcomes have been achieved using CAR-T cell therapy for haematological malignancies. Despite encouraging results, several challenges still pose eminent hurdles before being fully recognized. Directing CAR-T cells to target a single tumour associated antigen (TAA) as the case in haematological malignancies might be much simpler than targeting the extensive inhibitory microenvironments associated with solid tumours. This review focuses on the basic principles involved in development of CAR-T cells, emphasizing the differences between humoral IgG, T-cell receptors, CAR and Fcγ-CR constructs. It also highlights the complex inhibitory network that is usually associated with solid tumours, and tackles recent advances in the clinical studies that have provided great hope for the future use of CAR-T cell immunotherapy. While current Fcγ-CR T cell immunotherapy is in pre-clinical stage, is expected to provide a sound therapeutic approach to add to existing classical chemo- and radio-therapeutic modalities.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35116, Egypt.
| | - Asma Althani
- Biomedical Research Center, Qatar University, Doha 2713, Qatar
| | - Thomas Caceci
- Biomedical Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
| | - Roberto Arriga
- Department of Systems Medicine, Endocrinology and Medical Oncology, University of Rome "Tor Vergata", Rome, Italy
| | - Tommaso Sconocchia
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | | | | | - Mario Roselli
- Department of Systems Medicine, Endocrinology and Medical Oncology, University of Rome "Tor Vergata", Rome, Italy
| | - Sara Caratelli
- Institute of Translational Pharmacology-CNR, Rome, Italy
| | | | | |
Collapse
|
17
|
Xing X, Zou Z, He C, Hu Z, Liang K, Liang W, Wang Y, Du X. Enhanced antitumor effect of cytotoxic T lymphocytes induced by dendritic cells pulsed with colorectal cancer cell lysate expressing α-Gal epitopes. Oncol Lett 2019; 18:864-871. [PMID: 31289564 DOI: 10.3892/ol.2019.10376] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 04/24/2019] [Indexed: 01/08/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common types of gastrointestinal malignancy. Traditional therapeutic options for CRC exhibit a limited effect. Adoptive cellular therapy has emerged as a new treatment strategy for CRC. Dendritic cells (DCs) are potent antigen-presenting cells. Specific cytotoxic T lymphocytes (CTLs) activated by DCs pulsed with tumor lysate have been reported to be a safe and promising treatment approach for CRC. However, the antitumor effect of specific CTLs remains limited. The low immunogenicity of tumor-associated antigens (TAAs) is the main reason for this limited therapeutic effect. In the present study, α-gal epitopes were synthesized on the CRC cell line SW620 to increase the immunogenicity of TAAs. DCs were pulsed with α-gal-expressing tumor lysate and CTLs were activated by these DCs. The cytotoxicity of CTLs was measured in vitro. The results demonstrated that DCs pulsed with α-gal-expressing tumor lysate can increase the frequency of CD3+CD8+ CTLs and natural killer T cells, increase the level of tumor necrosis factor-α produced by CTLs and enhance the cytotoxicity of CTLs against tumor cells. Therefore, this novel approach may be an effective treatment strategy for patients with CRC.
Collapse
Affiliation(s)
- Xiaowei Xing
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Zhenyu Zou
- Department of Hernia and Abdominal Wall Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100853, P.R. China
| | - Changzheng He
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Zilong Hu
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Kai Liang
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Wentao Liang
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Yufeng Wang
- Department of Patient Admission Management, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Xiaohui Du
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
18
|
What is the Role of Hematopoietic Cell Transplantation (HCT) for Pediatric Acute Lymphoblastic Leukemia (ALL) in the Age of Chimeric Antigen Receptor T-Cell (CART) Therapy? J Pediatr Hematol Oncol 2019; 41:337-344. [PMID: 30973486 DOI: 10.1097/mph.0000000000001479] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
CD19 chimeric antigen receptor T-cell (CART) therapy has revolutionized the treatment of patients with relapsed/refractory hematologic malignancies, especially B-cell acute lymphoblastic leukemia. As CART immunotherapy expands from clinical trials to FDA-approved treatments, a consensus among oncologists and hematopoietic cell transplant (HCT) physicians is needed to identify which patients may benefit from consolidative HCT post-CART therapy. Here, we review CD19 CART therapy and the outcomes of published clinical trials, highlighting the use of post-CART HCT and the pattern of relapse after CD19 CART. At this time, the limited available long-term data from clinical trials precludes us from making definitive HCT recommendations. However, based on currently available data, we propose that consolidative HCT post-CART therapy be considered for all HCT-eligible patients and especially for pediatric patients with KMT2A-rearranged B-cell acute lymphoblastic leukemia.
Collapse
|
19
|
Schubert ML, Schmitt A, Sellner L, Neuber B, Kunz J, Wuchter P, Kunz A, Gern U, Michels B, Hofmann S, Hückelhoven-Krauss A, Kulozik A, Ho AD, Müller-Tidow C, Dreger P, Schmitt M. Treatment of patients with relapsed or refractory CD19+ lymphoid disease with T lymphocytes transduced by RV-SFG.CD19.CD28.4-1BBzeta retroviral vector: a unicentre phase I/II clinical trial protocol. BMJ Open 2019; 9:e026644. [PMID: 31110096 PMCID: PMC6530404 DOI: 10.1136/bmjopen-2018-026644] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION Chimeric antigen receptor (CAR) T cells spark hope for patients with CD19+ B cell neoplasia, including relapsed or refractory (r/r) acute lymphoblastic leukaemia (ALL) or r/r non-Hodgkin's lymphoma (NHL). Published studies have mostly used second-generation CARs with 4-1BB or CD28 as costimulatory domains. Preclinical results of third-generation CARs incorporating both elements have shown superiority concerning longevity and proliferation. The University Hospital of Heidelberg is the first institution to run an investigator-initiated trial (IIT) CAR T cell trial (Heidelberg Chimeric Antigen Receptor T cell Trial number 1 [HD-CAR-1]) in Germany with third-generation CD19-directed CAR T cells. METHODS AND ANALYSIS Adult patients with r/r ALL (stratum I), r/r NHL including chronic lymphocytic leukaemia, diffuse large B-cell lymphoma, follicular lymphoma or mantle cell lymphoma (stratum II) as well as paediatric patients with r/r ALL (stratum III) will be treated with autologous T-lymphocytes transduced by third-generation RV-SFG.CD19.CD28.4-1BB zeta retroviral vector (CD19.CAR T cells). The main purpose of this study is to evaluate safety and feasibility of escalating CD19.CAR T cell doses (1-20×106 transduced cells/m2) after lymphodepletion with fludarabine (flu) and cyclophosphamide (cyc). Patients will be monitored for cytokine release syndrome (CRS), neurotoxicity, i.e. CAR-T-cell-related encephalopathy syndrome (CRES) and/or other toxicities (primary objectives). Secondary objectives include evaluation of in vivo function and survival of CD19.CAR T cells and assessment of CD19.CAR T cell antitumour efficacy.HD-CAR-1 as a prospective, monocentric trial aims to make CAR T cell therapy accessible to patients in Europe. Currently, HD-CAR-1 is the first and only CAR T cell IIT in Germany. A third-generation Good Manufacturing Practice (GMP) grade retroviral vector, a broad spectrum of NHL, treatment of paediatric and adult ALL patients and inclusion of patients even after allogeneic stem cell transplantation (alloSCT) make this trial unique. ETHICS AND DISSEMINATION Ethical approval and approvals from the local and federal competent authorities were granted. Trial results will be reported via peer-reviewed journals and presented at conferences and scientific meetings. TRIAL REGISTRATION NUMBER Eudra CT 2016-004808-60; NCT03676504; Pre-results.
Collapse
Affiliation(s)
- Maria-Luisa Schubert
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Leopold Sellner
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK), National Centre for Tumour Diseases (NCT), Heidelberg, Germany
| | - Brigitte Neuber
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Joachim Kunz
- Department of Pediatric Hematology, Oncology and Immunology, Children’s Hospital, Heidelberg University Hospital, Heidelberg, Germany
| | - Patrick Wuchter
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alexander Kunz
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Ulrike Gern
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Birgit Michels
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Susanne Hofmann
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Andreas Kulozik
- Department of Pediatric Hematology, Oncology and Immunology, Children’s Hospital, Heidelberg University Hospital, Heidelberg, Germany
| | - Anthony D. Ho
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK), National Centre for Tumour Diseases (NCT), Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK), National Centre for Tumour Diseases (NCT), Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK), National Centre for Tumour Diseases (NCT), Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK), National Centre for Tumour Diseases (NCT), Heidelberg, Germany
| |
Collapse
|
20
|
Tumor-Specific Reactive Oxygen Species Accelerators Improve Chimeric Antigen Receptor T Cell Therapy in B Cell Malignancies. Int J Mol Sci 2019; 20:ijms20102469. [PMID: 31109083 PMCID: PMC6566309 DOI: 10.3390/ijms20102469] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/13/2019] [Accepted: 05/16/2019] [Indexed: 01/16/2023] Open
Abstract
Chimeric antigen receptor T cell (CART) therapy is currently one of the most promising treatment approaches in cancer immunotherapy. However, the immunosuppressive nature of the tumor microenvironment, in particular increased reactive oxygen species (ROS) levels, provides considerable limitations. In this study, we aimed to exploit increased ROS levels in the tumor microenvironment with prodrugs of ROS accelerators, which are specifically activated in cancer cells. Upon activation, ROS accelerators induce further generation of ROS. This leads to an accumulation of ROS in tumor cells. We hypothesized that the latter cells will be more susceptible to CARTs. CD19-specific CARTs were generated with a CD19.CAR.CD28.CD137zeta third-generation retroviral vector. Cytotoxicity was determined by chromium-51 release assay. Influence of the ROS accelerators on viability and phenotype of CARTs was determined by flow cytometry. The combination of CARTs with the ROS accelerator PipFcB significantly increased their cytotoxicity in the Burkitt lymphoma cell lines Raji and Daudi, as well as primary chronic lymphocytic leukemia cells. Exposure of CARTs to PipFcB for 48 h did not influence T cell exhaustion, viability, or T cell subpopulations. In summary, the combination of CARTs with ROS accelerators may improve adoptive immunotherapy and help to overcome tumor microenvironment-mediated treatment resistance.
Collapse
|
21
|
Stock S, Übelhart R, Schubert ML, Fan F, He B, Hoffmann JM, Wang L, Wang S, Gong W, Neuber B, Hückelhoven-Krauss A, Gern U, Christ C, Hexel M, Schmitt A, Schmidt P, Krauss J, Jäger D, Müller-Tidow C, Dreger P, Schmitt M, Sellner L. Idelalisib for optimized CD19-specific chimeric antigen receptor T cells in chronic lymphocytic leukemia patients. Int J Cancer 2019; 145:1312-1324. [PMID: 30737788 DOI: 10.1002/ijc.32201] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/21/2019] [Accepted: 01/28/2019] [Indexed: 12/14/2022]
Abstract
Despite encouraging results with chimeric antigen receptor T (CART) cells, outcome can still be improved by optimization of the CART cell generation process. The proportion of less-differentiated T cells within the transfused product is linked to enhanced in vivo CART cell expansion and long-term persistence. The clinically approved PI3Kδ inhibitor idelalisib is well established in the treatment of B cell malignancies. Besides B cell receptor pathway inhibition, idelalisib can modulate T cell differentiation and function. Here, detailed longitudinal analysis of idelalisib-induced effects on T cell phenotype and function was performed during CART cell production. A third generation CD19.CAR.CD28.CD137zeta CAR vector system was used. CART cells were generated from peripheral blood mononuclear cells of healthy donors (HDs) and chronic lymphocytic leukemia (CLL) patients. Idelalisib-based CART cell generation resulted in an enrichment of less-differentiated naïve-like T cells (CD45RA+CCR7+), decreased expression of the exhaustion markers PD-1 and Tim-3, as well as upregulation of the lymph node homing marker CD62L. Idelalisib increased transduction efficiency, but did not impair viability and cell expansion. Strikingly, CD4:CD8 ratios that were altered in CART cells from CLL patients were approximated to ratios in HDs by idelalisib. Furthermore, in vivo efficacy of idelalisib-treated CART cells was validated in a xenograft mouse model. Intracellular TNF-α and IFN-γ production decreased in presence of idelalisib. This effect was reversible after resting CART cells without idelalisib. In summary, PI3Kδ inhibition with idelalisib can improve CART cell products, particularly when derived from CLL patients. Further studies with idelalisib-based CART cell generation protocols are warranted.
Collapse
Affiliation(s)
- Sophia Stock
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Rudolf Übelhart
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit "Applied Tumor-Immunity", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Maria-Luisa Schubert
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Fuli Fan
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Bailin He
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Jean-Marc Hoffmann
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Lei Wang
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Sanmei Wang
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Wenjie Gong
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Brigitte Neuber
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Ulrike Gern
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Christiane Christ
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany
| | - Monika Hexel
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Patrick Schmidt
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Jürgen Krauss
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Leopold Sellner
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
22
|
Stock S, Hoffmann JM, Schubert ML, Wang L, Wang S, Gong W, Neuber B, Gern U, Schmitt A, Müller-Tidow C, Dreger P, Schmitt M, Sellner L. Influence of Retronectin-Mediated T-Cell Activation on Expansion and Phenotype of CD19-Specific Chimeric Antigen Receptor T Cells. Hum Gene Ther 2018; 29:1167-1182. [PMID: 30024314 DOI: 10.1089/hum.2017.237] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Enhanced in vivo expansion, long-term persistence of chimeric antigen receptor T (CART) cells, and efficient tumor eradication through these cells are linked to the proportion of less-differentiated cells in the CART cell product. Retronectin is well established as an adjuvant for improved retroviral transduction, while its property to enrich less-differentiated T cells is less known. In order to increase these subsets, this study investigated the effects of retronectin-mediated T-cell activation for CD19-specific CART cell production. Peripheral blood mononuclear cells of healthy donors and untreated chronic lymphocytic leukemia (CLL) patients without or with positive selection for CD3+ T cells were transduced with a CD19.CAR.CD28.CD137zeta third-generation retroviral vector. Activation of peripheral blood mononuclear cells was performed by CD3/CD28, CD3/CD28/retronectin, or CD3/retronectin. Interleukin-7 and -15 were supplemented to all cultures. Retronectin was used in all three activation protocols for retroviral transduction. Expansion was assessed by trypan blue staining. Viability, transduction efficiency, immune phenotype, and cytokine production were longitudinally analyzed by flow cytometry. Cytotoxic capacity of generated CART cells was evaluated using a classical chromium-51 release assay. Retronectin-mediated activation resulted in an enrichment of CD8+ cytotoxic CART cells and less-differentiated naïve-like T cells (CD45RA+CCR7+). Retronectin-activated CART cells showed increased cytotoxic activity. However, activation with retronectin decreased viability, expansion, transduction efficiency, and cytokine production, particularly of CLL patient-derived CART cells. Both retronectin-mediated activation protocols promoted a less-differentiated CART cell phenotype without comprising cytotoxic properties of healthy donor-derived CART cells. However, up-front retronectin resulted in reduced viability and expansion in CLL patients. This effect is probably attributed to the retronectin-mediated activation of B cells with prolonged CLL persistence. Consequently, CART cell expansion and generation failed. In summary, activation with retronectin should be performed with caution and may be limited to patients without a higher percentage of tumor cells in the peripheral blood.
Collapse
Affiliation(s)
- Sophia Stock
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany
| | - Jean-Marc Hoffmann
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany
| | - Maria-Luisa Schubert
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany
| | - Lei Wang
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany
| | - Sanmei Wang
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany
| | - Wenjie Gong
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany
| | - Brigitte Neuber
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany
| | - Ulrike Gern
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany
| | - Anita Schmitt
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany
| | - Carsten Müller-Tidow
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany .,2 National Center for Tumor Diseases , German Cancer Consortium, Heidelberg, Germany
| | - Peter Dreger
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany .,2 National Center for Tumor Diseases , German Cancer Consortium, Heidelberg, Germany
| | - Michael Schmitt
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany .,2 National Center for Tumor Diseases , German Cancer Consortium, Heidelberg, Germany
| | - Leopold Sellner
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany .,2 National Center for Tumor Diseases , German Cancer Consortium, Heidelberg, Germany
| |
Collapse
|
23
|
Alhakeem SS, McKenna MK, Oben KZ, Noothi SK, Rivas JR, Hildebrandt GC, Fleischman RA, Rangnekar VM, Muthusamy N, Bondada S. Chronic Lymphocytic Leukemia-Derived IL-10 Suppresses Antitumor Immunity. THE JOURNAL OF IMMUNOLOGY 2018; 200:4180-4189. [PMID: 29712773 PMCID: PMC6555426 DOI: 10.4049/jimmunol.1800241] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 04/06/2018] [Indexed: 12/21/2022]
Abstract
Chronic lymphocytic leukemia (CLL) patients progressively develop an immunosuppressive state. CLL patients have more plasma IL-10, an anti-inflammatory cytokine, than healthy controls. In vitro human CLL cells produce IL-10 in response to BCR cross-linking. We used the transgenic Eμ-T cell leukemia oncogene-1 (TCL1) mouse CLL model to study the role of IL-10 in CLL associated immunosuppression. Eμ-TCL mice spontaneously develop CLL because of a B cell-specific expression of the oncogene, TCL1. Eμ-TCL1 mouse CLL cells constitutively produce IL-10, which is further enhanced by BCR cross-linking, CLL-derived IL-10 did not directly affect survival of murine or human CLL cells in vitro. We tested the hypothesis that the CLL-derived IL-10 has a critical role in CLL disease in part by suppressing the host immune response to the CLL cells. In IL-10R-/- mice, wherein the host immune cells are unresponsive to IL-10-mediated suppressive effects, there was a significant reduction in CLL cell growth compared with wild type mice. IL-10 reduced the generation of effector CD4 and CD8 T cells. We also found that activation of BCR signaling regulated the production of IL-10 by both murine and human CLL cells. We identified the transcription factor, Sp1, as a novel regulator of IL-10 production by CLL cells and that it is regulated by BCR signaling via the Syk/MAPK pathway. Our results suggest that incorporation of IL-10 blocking agents may enhance current therapeutic regimens for CLL by potentiating host antitumor immune response.
Collapse
Affiliation(s)
- Sara S Alhakeem
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536.,Markey Cancer Center, University of Kentucky, Lexington, KY 40536
| | - Mary K McKenna
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536.,Markey Cancer Center, University of Kentucky, Lexington, KY 40536
| | - Karine Z Oben
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536.,Markey Cancer Center, University of Kentucky, Lexington, KY 40536
| | - Sunil K Noothi
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536.,Markey Cancer Center, University of Kentucky, Lexington, KY 40536.,Department of Radiation Medicine, University of Kentucky, Lexington, KY 40536
| | - Jacqueline R Rivas
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536.,Markey Cancer Center, University of Kentucky, Lexington, KY 40536
| | - Gerhard C Hildebrandt
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536.,Markey Cancer Center, University of Kentucky, Lexington, KY 40536.,Division of Hematology, Blood, and Marrow Transplantation, University of Kentucky, Lexington, KY 40536
| | - Roger A Fleischman
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536.,Division of Hematology, Blood, and Marrow Transplantation, University of Kentucky, Lexington, KY 40536
| | - Vivek M Rangnekar
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536.,Markey Cancer Center, University of Kentucky, Lexington, KY 40536.,Department of Radiation Medicine, University of Kentucky, Lexington, KY 40536
| | - Natarajan Muthusamy
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210; and.,Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Subbarao Bondada
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536; .,Markey Cancer Center, University of Kentucky, Lexington, KY 40536
| |
Collapse
|
24
|
Krackhardt AM, Anliker B, Hildebrandt M, Bachmann M, Eichmüller SB, Nettelbeck DM, Renner M, Uharek L, Willimsky G, Schmitt M, Wels WS, Schüssler-Lenz M. Clinical translation and regulatory aspects of CAR/TCR-based adoptive cell therapies-the German Cancer Consortium approach. Cancer Immunol Immunother 2018; 67:513-523. [PMID: 29380009 PMCID: PMC11028374 DOI: 10.1007/s00262-018-2119-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 01/20/2018] [Indexed: 12/17/2022]
Abstract
Adoptive transfer of T cells genetically modified by TCRs or CARs represents a highly attractive novel therapeutic strategy to treat malignant diseases. Various approaches for the development of such gene therapy medicinal products (GTMPs) have been initiated by scientists in recent years. To date, however, the number of clinical trials commenced in Germany and Europe is still low. Several hurdles may contribute to the delay in clinical translation of these therapeutic innovations including the significant complexity of manufacture and non-clinical testing of these novel medicinal products, the limited knowledge about the intricate regulatory requirements of the academic developers as well as limitations of funds for clinical testing. A suitable good manufacturing practice (GMP) environment is a key prerequisite and platform for the development, validation, and manufacture of such cell-based therapies, but may also represent a bottleneck for clinical translation. The German Cancer Consortium (DKTK) and the Paul-Ehrlich-Institut (PEI) have initiated joint efforts of researchers and regulators to facilitate and advance early phase, academia-driven clinical trials. Starting with a workshop held in 2016, stakeholders from academia and regulatory authorities in Germany have entered into continuing discussions on a diversity of scientific, manufacturing, and regulatory aspects, as well as the benefits and risks of clinical application of CAR/TCR-based cell therapies. This review summarizes the current state of discussions of this cooperative approach providing a basis for further policy-making and suitable modification of processes.
Collapse
Affiliation(s)
- Angela M Krackhardt
- Klinik und Poliklinik für Innere Medizin III, Hämatologie und Onkologie, Klinikum rechts der Isar, TU München, TUM School of Medicine, Munich, Germany.
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany.
| | - Brigitte Anliker
- Paul-Ehrlich-Institut (PEI, German Federal Institute for Vaccines and Biomedicines), Langen, Germany
| | - Martin Hildebrandt
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- TUMCells (Interdisciplinary Center for Cellular Therapies), TUM School of Medicine, Munich, Germany
| | - Michael Bachmann
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- Helmholtz Zentrum Dresden Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Radio and Tumorimmunology, Dresden, Germany
- Nationales Centrum für Tumorerkrankungen (NCT), Heidelberg and Dresden, Germany
| | - Stefan B Eichmüller
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- Nationales Centrum für Tumorerkrankungen (NCT), Heidelberg and Dresden, Germany
- GMP and T Cell Therapy Unit, DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
| | - Dirk M Nettelbeck
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
| | - Matthias Renner
- Paul-Ehrlich-Institut (PEI, German Federal Institute for Vaccines and Biomedicines), Langen, Germany
| | - Lutz Uharek
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- Stem Cell Facility, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Gerald Willimsky
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- Institute of Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Michael Schmitt
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- Department of Internal Medicine V, GMP Core Facility, Heidelberg University Hospital, Heidelberg, Germany
| | - Winfried S Wels
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Martina Schüssler-Lenz
- Paul-Ehrlich-Institut (PEI, German Federal Institute for Vaccines and Biomedicines), Langen, Germany
| |
Collapse
|
25
|
Ulrich P, Blaich G, Baumann A, Fagg R, Hey A, Kiessling A, Kronenberg S, Lindecrona RH, Mohl S, Richter WF, Tibbitts J, Crameri F, Weir L. Biotherapeutics in non-clinical development: Strengthening the interface between safety, pharmacokinetics-pharmacodynamics and manufacturing. Regul Toxicol Pharmacol 2018; 94:91-100. [PMID: 29355662 DOI: 10.1016/j.yrtph.2018.01.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 01/14/2018] [Indexed: 12/29/2022]
Abstract
Biological drugs comprise a wide field of different modalities with respect to structure, pharmacokinetics and pharmacological function. Considerable non-clinical experience in the development of proteins (e.g. insulin) and antibodies has been accumulated over the past thirty years. In order to improve the efficacy and the safety of these biotherapeutics, Fc modifications (e.g. Fc silent antibody versions), combinations (antibody-drug conjugates, protein-nanoparticle combinations), and new constructs (darpins, fynomers) have been introduced. In the last decade, advanced therapy medicinal products (ATMPs) in research and development have become a considerable and strongly growing part of the biotherapeutic portfolio. ATMPs consisting of gene and cell therapy modalities or even combinations of them, further expand the level of complexity, which already exists in non-clinical development strategies for biological drugs and has thereby led to a further diversification of expertise in safety and PKPD assessment of biological drugs. It is the fundamental rationale of the BioSafe meetings, held yearly in the EU and in the US, to convene experts on a regular basis and foster knowledge exchange and mutual understanding in this fast growing area. In order to reflect at least partially the variety of the biotherapeutics field, the 2016 EU BioSafe meeting addressed the following topics in six sessions: (i) In vitro Meets in vivo to Leverage Biologics Development (ii) New developments and regulatory considerations in the cell and gene therapy field (iii) CMC Challenges with Biologics development (iv) Minipigs in non-clinical safety assessment (v) Opportunities of PKPD Assessment in Less Common Administration Routes In the breakout sessions the following questions were discussed: (i) Cynomolgus monkey as a reprotoxicology Species: Impact of Immunomodulators on Early Pregnancy Maintenance (ii) Safety Risk of Inflammation and Autoimmunity Induced by Immunomodulators (iii) Experience with non-GMP Material in Pivotal Non-clinical Safety Studies to Support First in Man (FiM) Trials (iv) Safety Assessment of Combination Products for Non-oncology.
Collapse
Affiliation(s)
| | | | | | | | - Adam Hey
- Novartis Pharma, Basel, Switzerland
| | | | - Sven Kronenberg
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | | | - Silke Mohl
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | - Wolfgang F Richter
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | | | - Flavio Crameri
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | | |
Collapse
|
26
|
Hoffmann JM, Schubert ML, Wang L, Hückelhoven A, Sellner L, Stock S, Schmitt A, Kleist C, Gern U, Loskog A, Wuchter P, Hofmann S, Ho AD, Müller-Tidow C, Dreger P, Schmitt M. Differences in Expansion Potential of Naive Chimeric Antigen Receptor T Cells from Healthy Donors and Untreated Chronic Lymphocytic Leukemia Patients. Front Immunol 2018; 8:1956. [PMID: 29375575 PMCID: PMC5767585 DOI: 10.3389/fimmu.2017.01956] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/19/2017] [Indexed: 12/20/2022] Open
Abstract
Introduction Therapy with chimeric antigen receptor T (CART) cells for hematological malignancies has shown promising results. Effectiveness of CART cells may depend on the ratio of naive (TN) vs. effector (TE) T cells, TN cells being responsible for an enduring antitumor activity through maturation. Therefore, we investigated factors influencing the TN/TE ratio of CART cells. Materials and methods CART cells were generated upon transduction of peripheral blood mononuclear cells with a CD19.CAR-CD28-CD137zeta third generation retroviral vector under two different stimulating culture conditions: anti-CD3/anti-CD28 antibodies adding either interleukin (IL)-7/IL-15 or IL-2. CART cells were maintained in culture for 20 days. We evaluated 24 healthy donors (HDs) and 11 patients with chronic lymphocytic leukemia (CLL) for the composition of cell subsets and produced CART cells. Phenotype and functionality were tested using flow cytometry and chromium release assays. Results IL-7/IL-15 preferentially induced differentiation into TN, stem cell memory (TSCM: naive CD27+ CD95+), CD4+ and CXCR3+ CART cells, while IL-2 increased effector memory (TEM), CD56+ and CD4+ T regulatory (TReg) CART cells. The net amplification of different CART subpopulations derived from HDs and untreated CLL patients was compared. Particularly the expansion of CD4+ CARTN cells differed significantly between the two groups. For HDs, this subtype expanded >60-fold, whereas CD4+ CARTN cells of untreated CLL patients expanded less than 10-fold. Expression of exhaustion marker programmed cell death 1 on CARTN cells on day 10 of culture was significantly higher in patient samples compared to HD samples. As the percentage of malignant B cells was expectedly higher within patient samples, an excessive amount of B cells during culture could account for the reduced expansion potential of CARTN cells in untreated CLL patients. Final TN/TE ratio stayed <0.3 despite stimulation condition for patients, whereas this ratio was >2 in samples from HDs stimulated with IL-7/IL-15, thus demonstrating efficient CARTN expansion. Conclusion Untreated CLL patients might constitute a challenge for long-lasting CART effects in vivo since only a low number of TN among the CART product could be generated. Depletion of malignant B cells before starting CART production might be considered to increase the TN/TE ratio within the CART product.
Collapse
Affiliation(s)
- Jean-Marc Hoffmann
- Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Maria-Luisa Schubert
- Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Lei Wang
- Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Angela Hückelhoven
- Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Leopold Sellner
- Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Sophia Stock
- Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Anita Schmitt
- Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Christian Kleist
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Ulrike Gern
- Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Angelica Loskog
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Patrick Wuchter
- Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
| | - Susanne Hofmann
- Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Anthony D Ho
- Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Carsten Müller-Tidow
- Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Peter Dreger
- Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Michael Schmitt
- Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany
| |
Collapse
|
27
|
Raikar SS, Fleischer LC, Moot R, Fedanov A, Paik NY, Knight KA, Doering CB, Spencer HT. Development of chimeric antigen receptors targeting T-cell malignancies using two structurally different anti-CD5 antigen binding domains in NK and CRISPR-edited T cell lines. Oncoimmunology 2017; 7:e1407898. [PMID: 29399409 DOI: 10.1080/2162402x.2017.1407898] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 10/29/2017] [Accepted: 11/06/2017] [Indexed: 12/21/2022] Open
Abstract
Relapsed T-cell malignancies have poor outcomes when treated with chemotherapy, but survival after allogeneic bone marrow transplantation (BMT) approaches 50%. A limitation to BMT is the difficulty of achieving remission prior to transplant. Chimeric antigen receptor (CAR) T-cell therapy has shown successes in B-cell malignancies. This approach is difficult to adapt for the treatment of T-cell disease due to lack of a T-lymphoblast specific antigen and the fratricide of CAR T cells that occurs with T-cell antigen targeting. To circumvent this problem two approaches were investigated. First, a natural killer (NK) cell line, which does not express CD5, was used for CAR expression. Second, CRISPR-Cas9 genome editing technology was used to knockout CD5 expression in CD5-positive Jurkat T cells and in primary T cells, allowing for the use of CD5-negative T cells for CAR expression. Two structurally distinct anti-CD5 sequences were also tested, i) a traditional immunoglobulin-based single chain variable fragment (scFv) and ii) a lamprey-derived variable lymphocyte receptor (VLR), which we previously showed can be used for CAR-based recognition. Our results show i) both CARs yield comparable T-cell activation and NK cell-based cytotoxicity when targeting CD5-positive cells, ii) CD5-edited CAR-modified Jurkat T cells have reduced self-activation compared to that of CD5-positive CAR-modified T cells, iii) CD5-edited CAR-modified Jurkat T cells have increased activation in the presence of CD5-positive target cells compared to that of CD5-positive CAR-modified T cells, and iv) although modest effects were seen, a mouse model using the CAR-expressing NK cell line showed the scFv-CAR was superior to the VLR-CAR in delaying disease progression.
Collapse
Affiliation(s)
- Sunil S Raikar
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Lauren C Fleischer
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.,Department of Molecular and Systems Pharmacology, Graduate Division of Biological and Biomedical Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Robert Moot
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.,Department of Molecular and Systems Pharmacology, Graduate Division of Biological and Biomedical Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Andrew Fedanov
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Na Yoon Paik
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Kristopher A Knight
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Christopher B Doering
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.,Department of Molecular and Systems Pharmacology, Graduate Division of Biological and Biomedical Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - H Trent Spencer
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.,Department of Molecular and Systems Pharmacology, Graduate Division of Biological and Biomedical Sciences, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
28
|
Schubert ML, Hoffmann JM, Dreger P, Müller-Tidow C, Schmitt M. Chimeric antigen receptor transduced T cells: Tuning up for the next generation. Int J Cancer 2017; 142:1738-1747. [PMID: 29119551 DOI: 10.1002/ijc.31147] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/05/2017] [Accepted: 10/23/2017] [Indexed: 12/17/2022]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has recently achieved impressive clinical outcome in patients with CD19-positive hematologic malignancies. Extrapolation of CAR T cell treatment to solid tumors, however, has not yet yielded similar results. This might be due to intrinsic causes, e.g. insufficient CAR T cell activation or CAR toxicity as well as extrinsic factors displaying an unfavorable tumor environment for CAR T cells by raising physical and chemical barriers. In this review, we discuss the advantages as well as major obstacles of CAR T cell therapy, particularly in the context of solid tumors, and focus on efforts and novel strategies in CAR T cell development.
Collapse
Affiliation(s)
- Maria-Luisa Schubert
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Jean-Marc Hoffmann
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| |
Collapse
|
29
|
Eltoukhy HS, Sinha G, Moore CA, Sandiford OA, Rameshwar P. Immune modulation by a cellular network of mesenchymal stem cells and breast cancer cell subsets: Implication for cancer therapy. Cell Immunol 2017; 326:33-41. [PMID: 28779846 DOI: 10.1016/j.cellimm.2017.07.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 07/28/2017] [Accepted: 07/29/2017] [Indexed: 02/07/2023]
Abstract
The immune modulatory properties of mesenchymal stem cells (MSCs) are mostly controlled by the particular microenvironment. Cancer stem cells (CSCs), which can initiate a clinical tumor, have been the subject of intense research. This review article discusses investigative studies of the roles of MSCs on cancer biology including on CSCs, and the potential as drug delivery to tumors. An understanding of how MSCs behave in the tumor microenvironment to facilitate the survival of tumor cells would be crucial to identify drug targets. More importantly, since CSCs survive for decades in dormancy for later resurgence, studies are presented to show how MSCs could be involved in maintaining dormancy. Although the mechanism by which CSCs survive is complex, this article focus on the cellular involvement of MSCs with regard to immune responses. We discuss the immunomodulatory mechanisms of MSC-CSC interaction in the context of therapeutic outcomes in oncology. We also discuss immunotherapy as a potential to circumventing this immune modulation.
Collapse
Affiliation(s)
- Hussam S Eltoukhy
- Rutgers, New Jersey Medical School, Department of Medicine-Hematology-Oncology, Newark, NJ 07103, USA
| | - Garima Sinha
- Rutgers, New Jersey Medical School, Department of Medicine-Hematology-Oncology, Newark, NJ 07103, USA
| | - Caitlyn A Moore
- Rutgers, New Jersey Medical School, Department of Medicine-Hematology-Oncology, Newark, NJ 07103, USA
| | - Oleta A Sandiford
- Rutgers, New Jersey Medical School, Department of Medicine-Hematology-Oncology, Newark, NJ 07103, USA
| | - Pranela Rameshwar
- Rutgers, New Jersey Medical School, Department of Medicine-Hematology-Oncology, Newark, NJ 07103, USA.
| |
Collapse
|
30
|
Li H, Zhao Y. Increasing the safety and efficacy of chimeric antigen receptor T cell therapy. Protein Cell 2017; 8:573-589. [PMID: 28434147 PMCID: PMC5546931 DOI: 10.1007/s13238-017-0411-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/07/2017] [Indexed: 12/19/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy is a promising cancer treatment that has recently been undergoing rapid development. However, there are still some major challenges, including precise tumor targeting to avoid off-target or “on-target/off-tumor” toxicity, adequate T cell infiltration and migration to solid tumors and T cell proliferation and persistence across the physical and biochemical barriers of solid tumors. In this review, we focus on the primary challenges and strategies to design safe and effective CAR T cells, including using novel cutting-edge technologies for CAR and vector designs to increase both the safety and efficacy, further T cell modification to overcome the tumor-associated immune suppression, and using gene editing technologies to generate universal CAR T cells. All these efforts promote the development and evolution of CAR T cell therapy and move toward our ultimate goal—curing cancer with high safety, high efficacy, and low cost.
Collapse
MESH Headings
- Cell Movement/immunology
- Cell Proliferation
- Gene Expression
- Genetic Vectors/chemistry
- Genetic Vectors/metabolism
- Humans
- Immunotherapy, Adoptive/methods
- Lymphocyte Activation
- Lymphocytes, Tumor-Infiltrating/cytology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/transplantation
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/pathology
- Neoplasms/therapy
- Patient Safety
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Recombinant Fusion Proteins/chemistry
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Signal Transduction
- Single-Chain Antibodies/chemistry
- Single-Chain Antibodies/genetics
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Treatment Outcome
Collapse
Affiliation(s)
- Hua Li
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104-5156, USA
- Cancer Center, Chengdu Military General Hospital, Chengdu, 610083, China
| | - Yangbing Zhao
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104-5156, USA.
| |
Collapse
|
31
|
Yu S, Li A, Liu Q, Li T, Yuan X, Han X, Wu K. Chimeric antigen receptor T cells: a novel therapy for solid tumors. J Hematol Oncol 2017; 10:78. [PMID: 28356156 PMCID: PMC5372296 DOI: 10.1186/s13045-017-0444-9] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 03/16/2017] [Indexed: 12/20/2022] Open
Abstract
The chimeric antigen receptor T (CAR-T) cell therapy is a newly developed adoptive antitumor treatment. Theoretically, CAR-T cells can specifically localize and eliminate tumor cells by interacting with the tumor-associated antigens (TAAs) expressing on tumor cell surface. Current studies demonstrated that various TAAs could act as target antigens for CAR-T cells, for instance, the type III variant epidermal growth factor receptor (EGFRvIII) was considered as an ideal target for its aberrant expression on the cell surface of several tumor types. CAR-T cell therapy has achieved gratifying breakthrough in hematological malignancies and promising outcome in solid tumor as showed in various clinical trials. The third generation of CAR-T demonstrates increased antitumor cytotoxicity and persistence through modification of CAR structure. In this review, we summarized the preclinical and clinical progress of CAR-T cells targeting EGFR, human epidermal growth factor receptor 2 (HER2), and mesothelin (MSLN), as well as the challenges for CAR-T cell therapy.
Collapse
Affiliation(s)
- Shengnan Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Anping Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Qian Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tengfei Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xun Yuan
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
32
|
Nandagopal L, Mehta A. Treatment approaches of hard-to-treat non-Hodgkin lymphomas. Expert Rev Hematol 2017; 10:259-273. [DOI: 10.1080/17474086.2017.1283214] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
| | - Amitkumar Mehta
- Hematology & Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|