1
|
Mucha M, Štach M, Kaštánková I, Rychlá J, Vydra J, Lesný P, Otáhal P. Good manufacturing practice-grade generation of CD19 and CD123-specific CAR-T cells using piggyBac transposon and allogeneic feeder cells in patients diagnosed with B-cell non-Hodgkin lymphoma and acute myeloid leukemia. Front Immunol 2024; 15:1415328. [PMID: 39192973 PMCID: PMC11347927 DOI: 10.3389/fimmu.2024.1415328] [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: 04/10/2024] [Accepted: 07/18/2024] [Indexed: 08/29/2024] Open
Abstract
Background The non-viral production of CAR-T cells through electroporation of transposon DNA plasmids is an alternative approach to lentiviral/retroviral methods. This method is particularly suitable for early-phase clinical trials involving novel types of CAR-T cells. The primary disadvantage of non-viral methods is the lower production efficiency compared to viral-based methods, which becomes a limiting factor for CAR-T production, especially in chemotherapy-pretreated lymphopenic patients. Methods We describe a good manufacturing practice (GMP)-compliant protocol for producing CD19 and CD123-specific CAR-T cells based on the electroporation of transposon vectors. The lymphocytes were purified from the blood of patients undergoing chemotherapy for B-NHL or AML and were electroporated with piggyBac transposon encoding CAR19 or CAR123, respectively. Electroporated cells were then polyclonally activated by anti-CD3/CD28 antibodies and a combination of cytokines (IL-4, IL-7, IL-21). The expansion was carried out in the presence of irradiated allogeneic blood-derived mononuclear cells (i.e., the feeder) for up to 21 days. Results Expansion in the presence of the feeder enhanced CAR-T production yield (4.5-fold in CAR19 and 9.3-fold in CAR123). Detailed flow-cytometric analysis revealed the persistence of early-memory CAR-T cells and a low vector-copy number after production in the presence of the feeder, with no negative impact on the cytotoxicity of feeder-produced CAR19 and CAR123 T cells. Furthermore, large-scale manufacturing of CAR19 carried out under GMP conditions using PBMCs obtained from B-NHL patients (starting number=200x10e6 cells) enabled the production of >50x10e6 CAR19 in 7 out of 8 cases in the presence of the feeder while only in 2 out of 8 cases without the feeder. Conclusions The described approach enables GMP-compatible production of sufficient numbers of CAR19 and CAR123 T cells for clinical application and provides the basis for non-viral manufacturing of novel experimental CAR-T cells that can be tested in early-phase clinical trials. This manufacturing approach can complement and advance novel experimental immunotherapeutic strategies against human hematologic malignancies.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Antigens, CD19/immunology
- Antigens, CD19/genetics
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- DNA Transposable Elements
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/genetics
- Feeder Cells
- Lymphoma, B-Cell/therapy
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Electroporation
- Allogeneic Cells/immunology
Collapse
Affiliation(s)
- Martin Mucha
- Institute of Hematology and Blood Transfusion, Prague, Czechia
- Faculty of Science, Charles University, Prague, Czechia
| | - Martin Štach
- Institute of Hematology and Blood Transfusion, Prague, Czechia
- Faculty of Science, Charles University, Prague, Czechia
| | - Iva Kaštánková
- Institute of Hematology and Blood Transfusion, Prague, Czechia
| | - Jana Rychlá
- Institute of Hematology and Blood Transfusion, Prague, Czechia
| | - Jan Vydra
- Institute of Hematology and Blood Transfusion, Prague, Czechia
| | - Petr Lesný
- Institute of Hematology and Blood Transfusion, Prague, Czechia
| | - Pavel Otáhal
- Institute of Hematology and Blood Transfusion, Prague, Czechia
| |
Collapse
|
2
|
Wang M, Jia L, Dai X, Zhang X. Advanced strategies in improving the immunotherapeutic effect of CAR-T cell therapy. Mol Oncol 2024; 18:1821-1848. [PMID: 38456710 PMCID: PMC11306536 DOI: 10.1002/1878-0261.13621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 12/23/2023] [Accepted: 02/20/2024] [Indexed: 03/09/2024] Open
Abstract
Chimeric antigen receptor (CAR-T) cell therapy is a newly developed immunotherapy strategy and has achieved satisfactory outcomes in the treatment of hematological malignancies. However, some adverse effects related to CAR-T cell therapy have to be resolved before it is widely used in clinics as a cancer treatment. Furthermore, the application of CAR-T cell therapy in the treatment of solid tumors has been hampered by numerous limitations. Therefore, it is essential to explore novel strategies to improve the therapeutic effect of CAR-T cell therapy. In this review, we summarized the recently developed strategies aimed at optimizing the generation of CAR-T cells and improving the anti-tumor efficiency of CAR-T cell therapy. Furthermore, the discovery of new targets for CAR-T cell therapy and the combined treatment strategies of CAR-T cell therapy with chemotherapy, radiotherapy, cancer vaccines and nanomaterials are highlighted.
Collapse
Affiliation(s)
- Minmin Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationFirst Hospital of Jilin UniversityChangchunChina
- National‐Local Joint Engineering Laboratory of Animal Models for Human DiseaseFirst Hospital of Jilin UniversityChangchunChina
| | - Linzi Jia
- Department of General MedicineShanxi Province Cancer HospitalTaiyuanChina
| | - Xiangpeng Dai
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationFirst Hospital of Jilin UniversityChangchunChina
- National‐Local Joint Engineering Laboratory of Animal Models for Human DiseaseFirst Hospital of Jilin UniversityChangchunChina
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationFirst Hospital of Jilin UniversityChangchunChina
- National‐Local Joint Engineering Laboratory of Animal Models for Human DiseaseFirst Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
3
|
Chen Q, Sun Y, Li H. Application of CAR-T cell therapy targeting mesothelin in solid tumor treatment. Discov Oncol 2024; 15:289. [PMID: 39023820 PMCID: PMC11258118 DOI: 10.1007/s12672-024-01159-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T-cell therapy is one of the most effective immunotherapies. CAR-T-cell therapy has achieved great success in the treatment of hematological malignancies. However, due to the characteristics of solid malignant tumors, such as on-target effects, off-tumor toxicity, an immunosuppressive tumor microenvironment (TME), and insufficient trafficking, CAR-T-cell therapy for solid tumors is still in the exploration stage. Mesothelin (MSLN) is a molecule expressed on the surface of various solid malignant tumor cells that is suitable as a target of tumor cells with high MSLN expression for CAR-T-cell therapy. This paper briefly described the development of CAR-T cell therapy and the structural features of MSLN, and especially summarized the strategies of structure optimization of MSLN-targeting CAR-T-cells and the enhancement methods of MSLN-targeting CAR-T cell anti-tumor efficacy by summarizing some preclinical experiment and clinical trials. When considering MSLN-targeting CAR-T-cell therapy as an example, this paper summarizes the efforts made by researchers in CAR-T-cell therapy for solid tumors and summarizes feasible treatment plans by integrating the existing research results.
Collapse
Affiliation(s)
- Qiuhong Chen
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan, 250014, People's Republic of China
| | - Yang Sun
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan, 250014, People's Republic of China
| | - Hua Li
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan, 250014, People's Republic of China.
| |
Collapse
|
4
|
Fox TA, Booth C. Improving access to gene therapy for rare diseases. Dis Model Mech 2024; 17:dmm050623. [PMID: 38639083 PMCID: PMC11051979 DOI: 10.1242/dmm.050623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024] Open
Abstract
Effective gene therapy approaches have been developed for many rare diseases, including inborn errors of immunity and metabolism, haemoglobinopathies and inherited blindness. Despite successful pre-clinical and clinical results, these gene therapies are not widely available, primarily for non-medical reasons. Lack of commercial interest in therapies for ultra-rare diseases, costs of development and complex manufacturing processes required for advanced therapy medicinal products (ATMPs) are some of the main problems that are restricting access. The complexities and costs of navigating the regulatory environments in different jurisdictions for treatments that affect small numbers of patients is a problem unique to ATMPS for rare and ultra-rare diseases. In this Perspective, we outline some of the challenges and potential solutions that, we hope, will improve access to gene therapy for rare diseases.
Collapse
Affiliation(s)
- Thomas A. Fox
- UCL Institute of Immunity and Transplantation, University College London, London, NW3 2PP, United Kingdom
| | - Claire Booth
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, UCL, London WC1N 1EH, UK
| |
Collapse
|
5
|
Khodke P, Kumbhar BV. Engineered CAR-T cells: An immunotherapeutic approach for cancer treatment and beyond. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 140:157-198. [PMID: 38762269 DOI: 10.1016/bs.apcsb.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
Chimeric Antigen Receptor (CAR) T cell therapy is a type of adoptive immunotherapy that offers a promising avenue for enhancing cancer treatment since traditional cancer treatments like chemotherapy, surgery, and radiation therapy have proven insufficient in completely eradicating tumors, despite the relatively positive outcomes. It has been observed that CAR-T cell therapy has shown promising results in treating the majority of hematological malignancies but also have a wide scope for other cancer types. CAR is an extra receptor on the T-cell that helps to increase and accelerate tumor destruction by efficiently activating the immune system. It is made up of three domains, the ectodomain, transmembrane, and the endodomain. The ectodomain is essential for antigen recognition and binding, whereas the co-stimulatory signal is transduced by the endodomain. To date, the Food and Drug Administration (FDA) has granted approval for six CAR-T cell therapies. However, despite its remarkable success, CAR-T therapy is associated with numerous adverse events and has certain limitations. This chapter focuses on the structure and function of the CAR domain, various generations of CAR, and the process of CAR-T cell development, adverse effects, and challenges in CAR-T therapy. CAR-T cell therapy also has scopes in other disease conditions which include systemic lupus erythematosus, multiple sclerosis, and myocardial fibrosis, etc.
Collapse
Affiliation(s)
- Purva Khodke
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be University, Mumbai, India
| | - Bajarang Vasant Kumbhar
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be University, Mumbai, India.
| |
Collapse
|
6
|
Han J, Zhang B, Zheng S, Jiang Y, Zhang X, Mao K. The Progress and Prospects of Immune Cell Therapy for the Treatment of Cancer. Cell Transplant 2024; 33:9636897241231892. [PMID: 38433349 PMCID: PMC10913519 DOI: 10.1177/09636897241231892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 03/05/2024] Open
Abstract
Immune cell therapy as a revolutionary treatment modality, significantly transformed cancer care. It is a specialized form of immunotherapy that utilizes living immune cells as therapeutic reagents for the treatment of cancer. Unlike traditional drugs, cell therapies are considered "living drugs," and these products are currently customized and require advanced manufacturing techniques. Although chimeric antigen receptor (CAR)-T cell therapies have received tremendous attention in the industry regarding the treatment of hematologic malignancies, their effectiveness in treating solid tumors is often restricted, leading to the emergence of alternative immune cell therapies. Tumor-infiltrating lymphocytes (TIL) cell therapy, cytokine-induced killer (CIK) cell therapy, dendritic cell (DC) vaccines, and DC/CIK cell therapy are designed to use the body's natural defense mechanisms to target and eliminate cancer cells, and usually have fewer side effects or risks. On the other hand, cell therapies, such as chimeric antigen receptor-T (CAR-T) cell, T cell receptor (TCR)-T, chimeric antigen receptor-natural killer (CAR-NK), or CAR-macrophages (CAR-M) typically utilize either autologous stem cells, allogeneic or xenogeneic cells, or genetically modified cells, which require higher levels of manipulation and are considered high risk. These high-risk cell therapies typically hold special characteristics in tumor targeting and signal transduction, triggering new anti-tumor immune responses. Recently, significant advances have been achieved in both basic and clinical researches on anti-tumor mechanisms, cell therapy product designs, and technological innovations. With swift technological integration and a high innovation landscape, key future development directions have emerged. To meet the demands of cell therapy technological advancements in treating cancer, we comprehensively and systematically investigate the technological innovation and clinical progress of immune cell therapies in this study. Based on the therapeutic mechanisms and methodological features of immune cell therapies, we analyzed the main technical advantages and clinical transformation risks associated with these therapies. We also analyzed and forecasted the application prospects, providing references for relevant enterprises with the necessary information to make informed decisions regarding their R&D direction selection.
Collapse
Affiliation(s)
- Jia Han
- Shanghai Information Center for Life Sciences, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Bowen Zhang
- Shanghai Information Center for Life Sciences, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Senyu Zheng
- Shanghai Information Center for Life Sciences, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
- School of Natural and Computing Sciences, University of Aberdeen, Aberdeen, UK
| | - Yuan Jiang
- Shanghai Information Center for Life Sciences, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Xiaopeng Zhang
- Shanghai World Trade Organization Affairs Consultation Center, Shanghai, China
| | - Kaiyun Mao
- Shanghai Information Center for Life Sciences, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
7
|
Louie RHY, Cai C, Samir J, Singh M, Deveson IW, Ferguson JM, Amos TG, McGuire HM, Gowrishankar K, Adikari T, Balderas R, Bonomi M, Ruella M, Bishop D, Gottlieb D, Blyth E, Micklethwaite K, Luciani F. CAR + and CAR - T cells share a differentiation trajectory into an NK-like subset after CD19 CAR T cell infusion in patients with B cell malignancies. Nat Commun 2023; 14:7767. [PMID: 38012187 PMCID: PMC10682404 DOI: 10.1038/s41467-023-43656-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy is effective in treating B cell malignancies, but factors influencing the persistence of functional CAR+ T cells, such as product composition, patients' lymphodepletion, and immune reconstitution, are not well understood. To shed light on this issue, here we conduct a single-cell multi-omics analysis of transcriptional, clonal, and phenotypic profiles from pre- to 1-month post-infusion of CAR+ and CAR- T cells from patients from a CARTELL study (ACTRN12617001579381) who received a donor-derived 4-1BB CAR product targeting CD19. Following infusion, CAR+ T cells and CAR- T cells shows similar differentiation profiles with clonally expanded populations across heterogeneous phenotypes, demonstrating clonal lineages and phenotypic plasticity. We validate these findings in 31 patients with large B cell lymphoma treated with CD19 CAR T therapy. For these patients, we identify using longitudinal mass-cytometry data an association between NK-like subsets and clinical outcomes at 6 months with both CAR+ and CAR- T cells. These results suggest that non-CAR-derived signals can provide information about patients' immune recovery and be used as correlate of clinically relevant parameters.
Collapse
Affiliation(s)
- Raymond Hall Yip Louie
- School of Computer Science and Engineering, UNSW Sydney, Sydney, NSW, Australia
- Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, NSW, Australia
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Curtis Cai
- Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, NSW, Australia
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Jerome Samir
- Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, NSW, Australia
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Mandeep Singh
- Garvan Institute for Medical Research, Sydney, NSW, Australia
| | - Ira W Deveson
- Garvan Institute for Medical Research, Sydney, NSW, Australia
| | | | - Timothy G Amos
- Garvan Institute for Medical Research, Sydney, NSW, Australia
| | - Helen Marie McGuire
- Ramaciotti Facility for Human Systems Biology, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Infection, Immunity and Inflammation Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Kavitha Gowrishankar
- Blood Transplant and Cell Therapies Program, Department of Haematology, Westmead Hospital, Sydney, NSW, Australia
- Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Thiruni Adikari
- Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, NSW, Australia
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | | | - Martina Bonomi
- Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, NSW, Australia
- Department of Physics, University of Bologna, Bologna, Italy
| | - Marco Ruella
- Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - David Bishop
- Blood Transplant and Cell Therapies Program, Department of Haematology, Westmead Hospital, Sydney, NSW, Australia
- Westmead Institute for Medical Research, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - David Gottlieb
- Blood Transplant and Cell Therapies Program, Department of Haematology, Westmead Hospital, Sydney, NSW, Australia
- Westmead Institute for Medical Research, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Emily Blyth
- Blood Transplant and Cell Therapies Program, Department of Haematology, Westmead Hospital, Sydney, NSW, Australia
- Westmead Institute for Medical Research, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Kenneth Micklethwaite
- Blood Transplant and Cell Therapies Program, Department of Haematology, Westmead Hospital, Sydney, NSW, Australia
- Westmead Institute for Medical Research, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
- NSW Health Pathology Blood Transplant and Cell Therapies Laboratory - ICPMR Westmead, Sydney, NSW, Australia
| | - Fabio Luciani
- Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, NSW, Australia.
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia.
- Garvan Institute for Medical Research, Sydney, NSW, Australia.
| |
Collapse
|
8
|
Cappuzzello E, Vigolo E, D’Accardio G, Astori G, Rosato A, Sommaggio R. How can Cytokine-induced killer cells overcome CAR-T cell limits. Front Immunol 2023; 14:1229540. [PMID: 37675107 PMCID: PMC10477668 DOI: 10.3389/fimmu.2023.1229540] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/02/2023] [Indexed: 09/08/2023] Open
Abstract
The successful treatment of patients affected by B-cell malignancies with Chimeric Antigen Receptor (CAR)-T cells represented a breakthrough in the field of adoptive cell therapy (ACT). However, CAR-T therapy is not an option for every patient, and several needs remain unmet. In particular, the production of CAR-T cells is expensive, labor-intensive and logistically challenging; additionally, the toxicities deriving from CAR-T cells infusion, such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), have been documented extensively. Alternative cellular therapy products such as Cytokine-induced killer (CIK) cells have the potential to overcome some of these obstacles. CIK cells are a heterogeneous population of polyclonal CD3+CD56+ T cells with phenotypic and functional properties of NK cells. CIK cell cytotoxicity is exerted in a major histocompatibility complex (MHC)-unrestricted manner through the engagement of natural killer group 2 member D (NKG2D) molecules, against a wide range of hematological and solid tumors without the need for prior antigen exposure or priming. The foremost potential of CIK cells lies in the very limited ability to induce graft-versus-host disease (GvHD) reactions in the allogeneic setting. CIK cells are produced with a simple and extremely efficient expansion protocol, which leads to a massive expansion of effector cells and requires a lower financial commitment compared to CAR-T cells. Indeed, CAR-T manufacturing involves the engineering with expensive GMP-grade viral vectors in centralized manufacturing facilities, whereas CIK cell production is successfully performed in local academic GMP facilities, and CIK cell treatment is now licensed in many countries. Moreover, the toxicities observed for CAR-T cells are not present in CIK cell-treated patients, thus further reducing the costs associated with hospitalization and post-infusion monitoring of patients, and ultimately encouraging the delivery of cell therapies in the outpatient setting. This review aims to give an overview of the limitations of CAR-T cell therapy and outline how the use of CIK cells could overcome such drawbacks thanks to their unique features. We highlight the undeniable advantages of using CIK cells as a therapeutic product, underlying the opportunity for further research on the topic.
Collapse
Affiliation(s)
- Elisa Cappuzzello
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Emilia Vigolo
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Giulia D’Accardio
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Giuseppe Astori
- Advanced Cellular Therapy Laboratory, Department of Hematology, San Bortolo Hospital of Vicenza, Vicenza, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Roberta Sommaggio
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| |
Collapse
|
9
|
Hiramatsu H. Current status of CAR-T cell therapy for pediatric hematologic malignancies. Int J Clin Oncol 2023; 28:729-735. [PMID: 37154980 DOI: 10.1007/s10147-023-02346-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/18/2023] [Indexed: 05/10/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common cancer in the pediatric population, and the long-term survival can reach 90%. However, approximately, 20% of pediatric ALL patients experience relapse and require second-line chemotherapy. This is frequently followed by hematopoietic stem cell transplantation, which can cause long-term sequelae. Recent advances in immunotherapy, such as monoclonal antibody therapy and chimeric antigen receptor (CAR)-T cell therapy, have revolutionized the treatment of relapsed and refractory ALL. Anti-CD19 CAR-T cells successfully eliminate B cell malignancies such as ALL. Tisagenlecleucel (Kymriah®) is the first CAR-T cell immunotherapy approved by the FDA. CAR-T cell therapy can cause specific adverse events (AEs) such as cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome, which are defined and graded according to the consensus grading system and treated with supportive therapies along with tocilizumab and corticosteroids. Other AEs include prolonged bone marrow suppression and hypogammaglobulinemia. Severe AEs are less common in the real-world experience than in clinical trials, probably due to better management of the patient before and during CAR-T cell therapy. The biggest challenge in CAR-T cell therapy against ALL is relapse. A high tumor burden on infusion, early loss of B cell aplasia, and minimal residual disease positivity after CAR-T cell infusion are predictive of relapse. Consolidative stem cell transplantation may improve the long-term outcome. The success of CD19 CAR-T cell therapy against B cell malignancy prompted extensive research into the use of CAR-T cells against other hematologic malignancies such as T cell leukemia or myeloid leukemia.
Collapse
Affiliation(s)
- Hidefumi Hiramatsu
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto City, Japan.
| |
Collapse
|
10
|
Faeq MH, Al-Haideri M, Mohammad TAM, Gharebakhshi F, Marofi F, Tahmasebi S, Modaresahmadi S. CAR-modified immune cells as a rapidly evolving approach in the context of cancer immunotherapies. Med Oncol 2023; 40:155. [PMID: 37083979 PMCID: PMC10119530 DOI: 10.1007/s12032-023-02019-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 03/28/2023] [Indexed: 04/22/2023]
Abstract
Nowadays, one of the main challenges clinicians face is malignancies. Through the progression of technology in recent years, tumor nature and tumor microenvironment (TME) can be better understood. Because of immune system involvement in tumorigenesis and immune cell dysfunction in the tumor microenvironment, clinicians encounter significant challenges in patient treatment and normal function recovery. The tumor microenvironment can stop the development of tumor antigen-specific helper and cytotoxic T cells in the tumor invasion process. Tumors stimulate the production of proinflammatory and immunosuppressive factors and cells that inhibit immune responses. Despite the more successful outcomes, the current cancer therapeutic approaches, including surgery, chemotherapy, and radiotherapy, have not been effective enough for tumor eradication. Hence, developing new treatment strategies such as monoclonal antibodies, adaptive cell therapies, cancer vaccines, checkpoint inhibitors, and cytokines helps improve cancer treatment. Among adoptive cell therapies, the interaction between the immune system and malignancies and using molecular biology led to the development of chimeric antigen receptor (CAR) T cell therapy. CAR-modified immune cells are one of the modern cancer therapeutic methods with encouraging outcomes in most hematological and solid cancers. The current study aimed to discuss the structure, formation, subtypes, and application of CAR immune cells in hematologic malignancies and solid tumors.
Collapse
Affiliation(s)
- Mohammed Hikmat Faeq
- Student of General Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maysoon Al-Haideri
- Department of Physiotherapy, Cihan University-Erbil, Kurdistan Region, Erbil, Iraq
| | - Talar Ahmad Merza Mohammad
- Department of Pharmacology, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
| | - Farshad Gharebakhshi
- Department of Radiology, School of Medicine, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Safa Tahmasebi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Shadan Modaresahmadi
- Department of Immunology and Biotechnology, Texas Tech University Health Siences Center, Abilene, TX, USA
| |
Collapse
|
11
|
Yagyu S, Nakazawa Y. piggyBac-transposon-mediated CAR-T cells for the treatment of hematological and solid malignancies. Int J Clin Oncol 2023; 28:736-747. [PMID: 36859566 DOI: 10.1007/s10147-023-02319-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/17/2023] [Indexed: 03/03/2023]
Abstract
Since the introduction of the use of chimeric antigen receptor T-cell therapy (CAR-T therapy) dramatically changed the therapeutic strategy for B cell tumors, various CAR-T cell products have been developed and applied to myeloid and solid tumors. Although viral vectors have been widely used to produce genetically engineered T cells, advances in genetic engineering have led to the development of methods for producing non-viral, gene-modified CAR-T cells. Recent progress has revealed that non-viral CAR-T cells have a significant impact not only on the simplicity of the production process and the accessibility of non-viral vectors but also on the function of the cells themselves. In this review, we focus on piggyBac-transposon-based CAR-T cells among non-viral, gene-modified CAR-T cells and discuss their characteristics, preclinical development, and recent clinical applications.
Collapse
Affiliation(s)
- Shigeki Yagyu
- Innovative Research and Liaison Organization, Shinshu University, 3-1-1, Asahi, Matsumoto, Nagano, Japan. .,Center for Advanced Research of Gene and Cell Therapy, Shinshu University, 3-1-1, Asahi, Matsumoto, Nagano, Japan. .,Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan.
| | - Yozo Nakazawa
- Center for Advanced Research of Gene and Cell Therapy, Shinshu University, 3-1-1, Asahi, Matsumoto, Nagano, Japan.,Department of Pediatrics, Shinshu University School of Medicine, 3-1-1, Asahi, Matsumoto, Nagano, Japan.,Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1, Asahi, Matsumoto, Nagano, Japan
| |
Collapse
|
12
|
Moretti A, Ponzo M, Nicolette CA, Tcherepanova IY, Biondi A, Magnani CF. The Past, Present, and Future of Non-Viral CAR T Cells. Front Immunol 2022; 13:867013. [PMID: 35757746 PMCID: PMC9218214 DOI: 10.3389/fimmu.2022.867013] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/28/2022] [Indexed: 12/14/2022] Open
Abstract
Adoptive transfer of chimeric antigen receptor (CAR) T lymphocytes is a powerful technology that has revolutionized the way we conceive immunotherapy. The impressive clinical results of complete and prolonged response in refractory and relapsed diseases have shifted the landscape of treatment for hematological malignancies, particularly those of lymphoid origin, and opens up new possibilities for the treatment of solid neoplasms. However, the widening use of cell therapy is hampered by the accessibility to viral vectors that are commonly used for T cell transfection. In the era of messenger RNA (mRNA) vaccines and CRISPR/Cas (clustered regularly interspaced short palindromic repeat-CRISPR-associated) precise genome editing, novel and virus-free methods for T cell engineering are emerging as a more versatile, flexible, and sustainable alternative for next-generation CAR T cell manufacturing. Here, we discuss how the use of non-viral vectors can address some of the limitations of the viral methods of gene transfer and allow us to deliver genetic information in a stable, effective and straightforward manner. In particular, we address the main transposon systems such as Sleeping Beauty (SB) and piggyBac (PB), the utilization of mRNA, and innovative approaches of nanotechnology like Lipid-based and Polymer-based DNA nanocarriers and nanovectors. We also describe the most relevant preclinical data that have recently led to the use of non-viral gene therapy in emerging clinical trials, and the related safety and efficacy aspects. We will also provide practical considerations for future trials to enable successful and safe cell therapy with non-viral methods for CAR T cell generation.
Collapse
Affiliation(s)
- Alex Moretti
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione Monza e Brianza per il Bambino e la sua Mamma (MBBM), Monza, Italy
| | - Marianna Ponzo
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione Monza e Brianza per il Bambino e la sua Mamma (MBBM), Monza, Italy
| | | | | | - Andrea Biondi
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione Monza e Brianza per il Bambino e la sua Mamma (MBBM), Monza, Italy
- Department of Pediatrics, University of Milano - Bicocca, Milan, Italy
- Clinica Pediatrica, University of Milano - Bicocca/Fondazione MBBM, Monza, Italy
| | - Chiara F. Magnani
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione Monza e Brianza per il Bambino e la sua Mamma (MBBM), Monza, Italy
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Characterizing piggyBat-a transposase for genetic modification of T cells. Mol Ther Methods Clin Dev 2022; 25:250-263. [PMID: 35474955 PMCID: PMC9018555 DOI: 10.1016/j.omtm.2022.03.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 03/17/2022] [Indexed: 11/21/2022]
Abstract
Chimeric antigen receptor (CAR) T cells targeting CD19 have demonstrated remarkable efficacy in the treatment of B cell malignancies. Current CAR T cell manufacturing protocols are complex and costly due to their reliance on viral vectors. Non-viral systems of genetic modification, such as with transposase and transposon systems, offer a potential streamlined alternative for CAR T cell manufacture and are currently being evaluated in clinical trials. In this study, we utilized the previously described transposase from the little brown bat, designated piggyBat, for production of CD19-specific CAR T cells. PiggyBat demonstrates efficient CAR transgene delivery, with a relatively low variability in integration copy number across a range of manufacturing conditions as well as a similar integration site profile to super-piggyBac transposon and viral vectors. PiggyBat-generated CAR T cells demonstrate CD19-specific cytotoxic efficacy in vitro and in vivo. These data demonstrate that alternative, naturally occurring DNA transposons can be efficiently re-tooled to be exploited in real-world applications.
Collapse
|
14
|
Kamal-Bahl S, Puckett JT, Bagchi I, Miller-Sonet E, Huntington SF. Barriers and solutions to improve access for chimeric antigen receptor therapies. Immunotherapy 2022; 14:741-753. [PMID: 35621253 DOI: 10.2217/imt-2022-0037] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Chimeric antigen receptor T cells are a potentially curative new therapeutic option, but access challenges remain. The limited number of certified treatment centers and the need to travel to these centers, the expenses of travel and lodging and the out-of-pocket costs associated with treatment pose a challenge for patients. Further, the logistics of follow-up coupled with an ad hoc reimbursement environment make chimeric antigen receptor T-cell treatment an unattractive proposition for many providers. The patient-specific nature of these gene therapies has made scaling up production difficult for manufacturers. Providing expanded financial assistance for patients and education for community oncologists, and addressing reimbursement challenges, can alleviate some of these access barriers.
Collapse
Affiliation(s)
| | | | | | | | - Scott F Huntington
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
15
|
Suematsu M, Yagyu S, Nagao N, Kubota S, Shimizu Y, Tanaka M, Nakazawa Y, Imamura T. PiggyBac Transposon-Mediated CD19 Chimeric Antigen Receptor-T Cells Derived From CD45RA-Positive Peripheral Blood Mononuclear Cells Possess Potent and Sustained Antileukemic Function. Front Immunol 2022; 13:770132. [PMID: 35154098 PMCID: PMC8829551 DOI: 10.3389/fimmu.2022.770132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 01/05/2022] [Indexed: 12/13/2022] Open
Abstract
The quality of chimeric antigen receptor (CAR)-T cell products, namely, memory and exhaustion markers, affects the long-term functionality of CAR-T cells. We previously reported that piggyBac (PB) transposon-mediated CD19 CAR-T cells exhibit a memory-rich phenotype that is characterized by the high proportion of CD45RA+/C-C chemokine receptor type 7 (CCR7)+ T-cell fraction. To further investigate the favorable phenotype of PB-CD19 CAR-T cells, we generated PB-CD19 CAR-T cells from CD45RA+ and CD45RA− peripheral blood mononuclear cells (PBMCs) (RA+ CAR and RA− CAR, respectively), and compared their phenotypes and antitumor activity. RA+ CAR-T cells showed better transient gene transfer efficiency 24 h after transduction and superior expansion capacity after 14 days of culture than those shown by RA− CAR-T cells. RA+ CAR-T cells exhibited dominant CD8 expression, decreased expression of the exhaustion marker programmed cell death protein-1 (PD-1) and T-cell senescence marker CD57, and enriched naïve/stem cell memory fraction, which are associated with the longevity of CAR-T cells. Transcriptome analysis showed that canonical exhaustion markers were downregulated in RA+ CAR-T, even after antigen stimulation. Although antigen stimulation could increase CAR expression, leading to tonic CAR signaling and exhaustion, the expression of CAR molecules on cell surface after antigen stimulation in RA+ CAR-T cells was controlled at a relatively lower level than that in RA− CAR-T cells. In the in vivo stress test, RA+ CAR-T cells achieved prolonged tumor control with expansion of CAR-T cells compared with RA− CAR-T cells. CAR-T cells were not detected in the control or RA− CAR-T cells but RA+ CAR-T cells were expanded even after 50 days of treatment, as confirmed by sequential bone marrow aspiration. Our results suggest that PB-mediated RA+ CAR-T cells exhibit a memory-rich phenotype and superior antitumor function, thus CD45RA+ PBMCs might be considered an efficient starting material for PB-CAR-T cell manufacturing. This novel approach will be beneficial for effective treatment of B cell malignancies.
Collapse
Affiliation(s)
- Masaya Suematsu
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Shigeki Yagyu
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Nobuyoshi Nagao
- AGC Inc. Innovative Technology Laboratories, Yokohama, Japan
| | - Susumu Kubota
- AGC Inc. Materials Integration Laboratories, Yokohama, Japan
| | - Yuto Shimizu
- AGC Inc. Materials Integration Laboratories, Yokohama, Japan
| | - Miyuki Tanaka
- Department of Pediatrics, Shinshu University School of Medicine, Nagano, Japan
| | - Yozo Nakazawa
- Department of Pediatrics, Shinshu University School of Medicine, Nagano, Japan
| | - Toshihiko Imamura
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| |
Collapse
|
16
|
Gotti E, Tettamanti S, Zaninelli S, Cuofano C, Cattaneo I, Rotiroti MC, Cribioli S, Alzani R, Rambaldi A, Introna M, Golay J. Optimization of therapeutic T cell expansion in G-Rex device and applicability to large-scale production for clinical use. Cytotherapy 2022; 24:334-343. [DOI: 10.1016/j.jcyt.2021.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 02/02/2023]
|
17
|
Guha P, Katz SC. Strategies for manufacturing cell therapy products aligned with patient needs. Methods Cell Biol 2022; 167:203-226. [DOI: 10.1016/bs.mcb.2021.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
18
|
Micklethwaite KP, Gowrishankar K, Gloss BS, Li Z, Street JA, Moezzi L, Mach MA, Sutrave G, Clancy LE, Bishop DC, Louie RHY, Cai C, Foox J, MacKay M, Sedlazeck FJ, Blombery P, Mason CE, Luciani F, Gottlieb DJ, Blyth E. Investigation of product-derived lymphoma following infusion of piggyBac-modified CD19 chimeric antigen receptor T cells. Blood 2021; 138:1391-1405. [PMID: 33974080 PMCID: PMC8532197 DOI: 10.1182/blood.2021010858] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/24/2021] [Indexed: 11/20/2022] Open
Abstract
We performed a phase 1 clinical trial to evaluate outcomes in patients receiving donor-derived CD19-specific chimeric antigen receptor (CAR) T cells for B-cell malignancy that relapsed or persisted after matched related allogeneic hemopoietic stem cell transplant. To overcome the cost and transgene-capacity limitations of traditional viral vectors, CAR T cells were produced using the piggyBac transposon system of genetic modification. Following CAR T-cell infusion, 1 patient developed a gradually enlarging retroperitoneal tumor due to a CAR-expressing CD4+ T-cell lymphoma. Screening of other patients led to the detection, in an asymptomatic patient, of a second CAR T-cell tumor in thoracic para-aortic lymph nodes. Analysis of the first lymphoma showed a high transgene copy number, but no insertion into typical oncogenes. There were also structural changes such as altered genomic copy number and point mutations unrelated to the insertion sites. Transcriptome analysis showed transgene promoter-driven upregulation of transcription of surrounding regions despite insulator sequences surrounding the transgene. However, marked global changes in transcription predominantly correlated with gene copy number rather than insertion sites. In both patients, the CAR T-cell-derived lymphoma progressed and 1 patient died. We describe the first 2 cases of malignant lymphoma derived from CAR gene-modified T cells. Although CAR T cells have an enviable record of safety to date, our results emphasize the need for caution and regular follow-up of CAR T recipients, especially when novel methods of gene transfer are used to create genetically modified immune therapies. This trial was registered at www.anzctr.org.au as ACTRN12617001579381.
Collapse
MESH Headings
- Aged
- DNA Transposable Elements
- Gene Expression Regulation, Neoplastic
- Gene Transfer Techniques
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Leukemia, B-Cell/genetics
- Leukemia, B-Cell/therapy
- Lymphoma/etiology
- Lymphoma/genetics
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/therapy
- Male
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/therapeutic use
- T-Lymphocytes/metabolism
- Transcriptome
- Transgenes
Collapse
Affiliation(s)
- Kenneth P Micklethwaite
- Blood Transplant and Cell Therapies Program, Department of Haematology, Westmead Hospital, Sydney, NSW, Australia
- Blood Transplant and Cell Therapies Laboratory, NSW Health Pathology-ICPMR Westmead, Sydney, NSW, Australia
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Kavitha Gowrishankar
- Westmead Institute for Medical Research, Sydney, NSW, Australia
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Brian S Gloss
- Westmead Institute for Medical Research, Sydney, NSW, Australia
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Ziduo Li
- Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Janine A Street
- Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Leili Moezzi
- Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Melanie A Mach
- Westmead Institute for Medical Research, Sydney, NSW, Australia
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Gaurav Sutrave
- Blood Transplant and Cell Therapies Program, Department of Haematology, Westmead Hospital, Sydney, NSW, Australia
- Westmead Institute for Medical Research, Sydney, NSW, Australia
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Leighton E Clancy
- Blood Transplant and Cell Therapies Laboratory, NSW Health Pathology-ICPMR Westmead, Sydney, NSW, Australia
- Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - David C Bishop
- Blood Transplant and Cell Therapies Program, Department of Haematology, Westmead Hospital, Sydney, NSW, Australia
- Westmead Institute for Medical Research, Sydney, NSW, Australia
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Raymond H Y Louie
- Kirby Institute, University of New South Wales, Sydney. NSW, Australia
| | - Curtis Cai
- Kirby Institute, University of New South Wales, Sydney. NSW, Australia
| | - Jonathan Foox
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY
- The Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, New York, NY
| | - Matthew MacKay
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY
- The Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, New York, NY
| | - Fritz J Sedlazeck
- Human Genome Sequencing Center, College of Medicine, Baylor University, Houston, TX
| | - Piers Blombery
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Clinical Haematology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Christopher E Mason
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY
- The Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, New York, NY
- The Feil Family Brain and Mind Research Institute, New York, NY; and
- The WorldQuant Initiative for Quantitative Prediction, New York, NY
| | - Fabio Luciani
- Kirby Institute, University of New South Wales, Sydney. NSW, Australia
| | - David J Gottlieb
- Blood Transplant and Cell Therapies Program, Department of Haematology, Westmead Hospital, Sydney, NSW, Australia
- Westmead Institute for Medical Research, Sydney, NSW, Australia
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Emily Blyth
- Blood Transplant and Cell Therapies Program, Department of Haematology, Westmead Hospital, Sydney, NSW, Australia
- Blood Transplant and Cell Therapies Laboratory, NSW Health Pathology-ICPMR Westmead, Sydney, NSW, Australia
- Westmead Institute for Medical Research, Sydney, NSW, Australia
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
19
|
Li A, Kusuma GD, Driscoll D, Smith N, Wall DM, Levine BL, James D, Lim R. Advances in automated cell washing and concentration. Cytotherapy 2021; 23:774-786. [PMID: 34052112 DOI: 10.1016/j.jcyt.2021.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/16/2021] [Accepted: 04/05/2021] [Indexed: 02/01/2023]
Abstract
The successful commercialization of cell therapies requires thorough planning and consideration of product quality, cost and scale of the manufacturing process. The implementation of automation can be central to a robust and reproducible manufacturing process at industrialized scales. There have been a number of wash-and-concentrate devices developed for cell manufacturing. These technologies have arisen from transfusion medicine, hematopoietic stem cell and biologics manufacturing where operating mechanisms are distinct from manual centrifugation. This review describes the historical origin and fundamental technologies underlying each currently available wash-and-concentrate device as well as their relative advantages and disadvantages in cell therapy applications. Understanding the specific attributes and limitations of these technologies is essential to optimizing cell therapy manufacturing.
Collapse
Affiliation(s)
- Anqi Li
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia; Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Gina D Kusuma
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia; Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | | | | | - Dominic M Wall
- Cell Therapies Pty Ltd, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Bruce L Levine
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Rebecca Lim
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia; Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia.
| |
Collapse
|
20
|
Li C, Sun Y, Wang J, Tang L, Jiang H, Guo T, Liu L, Wu Y, Ai L, Xia L, Wu J, Lin Z, Qian Q, Hu Y, Mei H. PiggyBac-Generated CAR19-T Cells Plus Lenalidomide Cause Durable Complete Remission of Triple-Hit Refractory/Relapsed DLBCL: A Case Report. Front Immunol 2021; 12:599493. [PMID: 34113336 PMCID: PMC8186315 DOI: 10.3389/fimmu.2021.599493] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 03/18/2021] [Indexed: 11/13/2022] Open
Abstract
MYC/BCL2/BCL6 triple-hit lymphoma (THL) is an uncommon subset of high-grade B-cell lymphoma with aggressive clinical behavior and poor prognosis. TP53 mutation is an independently poor progonistic indicator in patients with THL, hence novel therapeutic strategies are needed for these patients. CD19-directed chimeric antigen receptor(CAR19)-T cell therapy has shown promising efficacy for relapsed/refractory diffuse large B cell lymphoma (RR DLBCL), but the majority of CAR19-T cell products to date have been manufactured using viral vectors. PiggyBac transposon system, with an inclination to memory T cells, offers a more convenient and economical alternative for transgene delivery. We herein report the first case of triple-hit RR DLBCL with TP53 mutation who was treated with piggyBac-generated CAR19-T cells and accompanied by grade 2 cytokine release syndrome. The patient obtained a complete remission (CR) in the 2nd month post-infusion and demanded maintenance therapy. Whether maintenance therapy is favorable and how to administrate it after CAR-T cell infusion remain controversial. Preclinical studies demonstrated that lenalidomide could enhance antitumor activity of CAR19-T cells. Therefore, we pioneered oral lenalidomide after CAR19-T therapy in the patient from the 4th month, and he discontinued after one cycle due to side effects. The patient has still kept sustained CR for over 24 months. Our case have firstly demonstrated the feasibility, preliminary safety and efficacy of piggyBac-produced CAR19-T cell therapy in triple-hit lymphoma. The innovative combination with lenalidomide warrants further investigation. Our findings shed new light on the possible solutions to improve short-term relapse after CAR19-T cell therapy in RR DLBCL. ChiCTR, number ChiCTR1800018111.
Collapse
Affiliation(s)
- Chenggong Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Yan Sun
- Shanghai Cell Therapy Group Co. Ltd., Shanghai, China
| | - Jing Wang
- Radiology Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Huiwen Jiang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Tao Guo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Lin Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaohui Wu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Lisha Ai
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Linghui Xia
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianjun Wu
- Shanghai Cell Therapy Group Co. Ltd., Shanghai, China
| | - Zhicai Lin
- Shanghai Cell Therapy Group Co. Ltd., Shanghai, China
| | - Qijun Qian
- Shanghai Cell Therapy Group Co. Ltd., Shanghai, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| |
Collapse
|
21
|
Phase I clinical trial of EGFR-specific CAR-T cells generated by the piggyBac transposon system in advanced relapsed/refractory non-small cell lung cancer patients. J Cancer Res Clin Oncol 2021; 147:3725-3734. [PMID: 34032893 DOI: 10.1007/s00432-021-03613-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/22/2021] [Indexed: 01/11/2023]
Abstract
PURPOSE This phase I clinical trial is designed to assess the safety and feasibility of the epidermal growth factor receptor (EGFR) chimeric antigen receptor (CAR) T-cell generated by the piggyBac transposon system in advanced relapsed/refractory non-small cell lung cancer (NSCLC) patients. Compared to viral systems, the piggyBac transposon system is a simpler, more economical, and alternative way to introduce chimeric antigen receptor (CAR) transgenes into T cells. METHODS This study recruited nine patients with advanced relapsed/refractory EGFR-positive NSCLC for two cycles of the piggyBac-generated EGFR-CAR T cells at dose of 1 × 106 cells/kg or 3 × 106 cells/kg of body weight. The patients were monitored for adverse events, clinical response, and persistence of plasma GFR-CAR T cells. RESULTS Infusions of piggyBac-generated EGFR-CAR T cells were well tolerated in all nine patients. The most common adverse events were grade 1 to 3 fever and there were no patients who experienced grade 4 adverse events or serious cytokine release syndrome. After treatment, eight of nine patients showed detectable EGFR-CAR T cells in their peripheral blood. One patient showed a partial response and lasted for more than 13 months, while six had stable disease, and two had progressed disease. The progression-free survival of these nine patients was 7.13 months (95% CI 2.71-17.10 months), while the median overall survival was 15.63 months (95% CI 8.82-22.03 months). CONCLUSION This Phase I clinical trial revealed that the non-viral piggyBac transposon system-engineered EGFR-CAR T-cell therapy is feasible and safe in treatment of EGFR-positive advanced relapsed/refractory NSCLC patients. Future study will assess it in more patients or even possibly with a higher dose. Trial registration number NCT03182816.
Collapse
|
22
|
Development of CAR T-cell lymphoma in two of ten patients effectively treated with piggyBac modified CD19 CAR T-cells. Blood 2021; 138:1504-1509. [PMID: 34010392 DOI: 10.1182/blood.2021010813] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/13/2021] [Indexed: 11/20/2022] Open
Abstract
CD19-specific chimeric antigen receptor (CAR19) T-cells effectively induce remission of B-cell malignancy, but the cost and complexity of production using viral vectors is a factor limiting widespread application. Furthermore, the small cargo capacity of viral vectors may hamper future development of more heavily engineered CAR T-cells. We demonstrated the feasibility of generating CAR19 T-cells from HLA-matched donors of sibling allogeneic hematopoietic stem cell transplant (HSCT) patients via a simple and inexpensive method using the high-capacity piggyBac transposon. A cohort of 10 patients with relapsed or refractory B-cell acute lymphoblastic leukemia or aggressive lymphoma following HSCT were the first human subjects to receive piggyBac-generated CAR19 T-cells. Treatment with intra-patient escalating doses of CAR19 T-cells was effective, with all 9 evaluable patients achieving complete remission. At a median follow-up of 18.0 months, 5 patients remained in complete remission of B-cell malignancy. One patient died of viral sepsis. Four patients developed cytokine release syndrome of maximum grade 2, and no neurotoxicity or new graft-versus-host disease occurred. However, two patients developed malignant CAR19 T-cell tumors, one of whom was successfully treated; one patient died of the secondary tumor. The piggyBac system represents a feasible alternative to viral vectors for the generation of effective CAR19 T-cells, but its oncogenic potential in the context of the described production process will need to be addressed before any further clinical use is possible. This trial was registered at www.anzctr.org.au as ACTRN12617001579381.
Collapse
|
23
|
Raes L, De Smedt SC, Raemdonck K, Braeckmans K. Non-viral transfection technologies for next-generation therapeutic T cell engineering. Biotechnol Adv 2021; 49:107760. [PMID: 33932532 DOI: 10.1016/j.biotechadv.2021.107760] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/24/2021] [Accepted: 04/24/2021] [Indexed: 12/24/2022]
Abstract
Genetically engineered T cells have sparked interest in advanced cancer treatment, reaching a milestone in 2017 with two FDA-approvals for CD19-directed chimeric antigen receptor (CAR) T cell therapeutics. It is becoming clear that the next generation of CAR T cell therapies will demand more complex engineering strategies and combinations thereof, including the use of revolutionary gene editing approaches. To date, manufacturing of CAR T cells mostly relies on γ-retroviral or lentiviral vectors, but their use is associated with several drawbacks, including safety issues, high manufacturing cost and vector capacity constraints. Non-viral approaches, including membrane permeabilization and carrier-based techniques, have therefore gained a lot of interest to replace viral transductions in the manufacturing of T cell therapeutics. This review provides an in-depth discussion on the avid search for alternatives to viral vectors, discusses key considerations for T cell engineering technologies, and provides an overview of the emerging spectrum of non-viral transfection technologies for T cells. Strengths and weaknesses of each technology will be discussed in relation to T cell engineering. Altogether, this work emphasizes the potential of non-viral transfection approaches to advance the next-generation of genetically engineered T cells.
Collapse
Affiliation(s)
- Laurens Raes
- Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Koen Raemdonck
- Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
24
|
Garcia-Aponte OF, Herwig C, Kozma B. Lymphocyte expansion in bioreactors: upgrading adoptive cell therapy. J Biol Eng 2021; 15:13. [PMID: 33849630 PMCID: PMC8042697 DOI: 10.1186/s13036-021-00264-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/29/2021] [Indexed: 12/25/2022] Open
Abstract
Bioreactors are essential tools for the development of efficient and high-quality cell therapy products. However, their application is far from full potential, holding several challenges when reconciling the complex biology of the cells to be expanded with the need for a manufacturing process that is able to control cell growth and functionality towards therapy affordability and opportunity. In this review, we discuss and compare current bioreactor technologies by performing a systematic analysis of the published data on automated lymphocyte expansion for adoptive cell therapy. We propose a set of requirements for bioreactor design and identify trends on the applicability of these technologies, highlighting the specific challenges and major advancements for each one of the current approaches of expansion along with the opportunities that lie in process intensification. We conclude on the necessity to develop targeted solutions specially tailored for the specific stimulation, supplementation and micro-environmental needs of lymphocytes’ cultures, and the benefit of applying knowledge-based tools for process control and predictability.
Collapse
Affiliation(s)
- Oscar Fabian Garcia-Aponte
- Research Area Biochemical Engineering, Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Gumpendorferstraße 1a, 1060, Vienna, Austria
| | - Christoph Herwig
- Research Area Biochemical Engineering, Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Gumpendorferstraße 1a, 1060, Vienna, Austria.
| | - Bence Kozma
- Research Area Biochemical Engineering, Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Gumpendorferstraße 1a, 1060, Vienna, Austria
| |
Collapse
|
25
|
Lukjanov V, Koutná I, Šimara P. CAR T-Cell Production Using Nonviral Approaches. J Immunol Res 2021; 2021:6644685. [PMID: 33855089 PMCID: PMC8019376 DOI: 10.1155/2021/6644685] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/30/2021] [Accepted: 03/19/2021] [Indexed: 01/01/2023] Open
Abstract
Chimeric antigen receptor T-cells (CAR T-cells) represent a novel and promising approach in cancer immunotherapy. According to the World Health Organization (WHO), the number of oncological patients is steadily growing in developed countries despite immense progress in oncological treatments, and the prognosis of individual patients is still relatively poor. Exceptional results have been recorded for CAR T-cell therapy in patients suffering from B-cell malignancies. This success opens up the possibility of using the same approach for other types of cancers. To date, the most common method for CAR T-cell generation is the use of viral vectors. However, dealing with virus-derived vectors brings possible obstacles in the CAR T-cell manufacturing process owing to strict regulations and high cost demands. Alternative approaches may facilitate further development and the transfer of the method to clinical practice. The most promising substitutes for virus-derived vectors are transposon-derived vectors, most commonly sleeping beauty, which offer great coding capability and a safe integration profile while maintaining a relatively low production cost. This review is aimed at summarizing the state of the art of nonviral approaches in CAR T-cell generation, with a unique perspective on the conditions in clinical applications and current Good Manufacturing Practice. If CAR T-cell therapy is to be routinely used in medical practice, the manufacturing cost and complexity need to be as low as possible, and transposon-based vectors seem to meet these criteria better than viral-based vectors.
Collapse
Affiliation(s)
- Viktor Lukjanov
- Masaryk University Brno, Faculty of Medicine, Department of Histology and Embryology, Kamenice 5, Brno 62500, Czech Republic
- St. Anne's University Hospital Brno, International Clinical Research Center, Pekarska 53, Brno 656 91, Czech Republic
| | - Irena Koutná
- Masaryk University Brno, Faculty of Medicine, Department of Histology and Embryology, Kamenice 5, Brno 62500, Czech Republic
- St. Anne's University Hospital Brno, International Clinical Research Center, Pekarska 53, Brno 656 91, Czech Republic
| | - Pavel Šimara
- Masaryk University Brno, Faculty of Medicine, Department of Histology and Embryology, Kamenice 5, Brno 62500, Czech Republic
- St. Anne's University Hospital Brno, International Clinical Research Center, Pekarska 53, Brno 656 91, Czech Republic
| |
Collapse
|
26
|
Akhoundi M, Mohammadi M, Sahraei SS, Sheykhhasan M, Fayazi N. CAR T cell therapy as a promising approach in cancer immunotherapy: challenges and opportunities. Cell Oncol (Dordr) 2021; 44:495-523. [PMID: 33759063 DOI: 10.1007/s13402-021-00593-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-modified T cell therapy has shown great potential in the immunotherapy of patients with hematologic malignancies. In spite of this striking achievement, there are still major challenges to overcome in CAR T cell therapy of solid tumors, including treatment-related toxicity and specificity. Also, other obstacles may be encountered in tackling solid tumors, such as their immunosuppressive microenvironment, the heterogeneous expression of cell surface markers, and the cumbersome arrival of T cells at the tumor site. Although several strategies have been developed to overcome these challenges, aditional research aimed at enhancing its efficacy with minimum side effects, the design of precise yet simplified work flows and the possibility to scale-up production with reduced costs and related risks is still warranted. CONCLUSIONS Here, we review main strategies to establish a balance between the toxicity and activity of CAR T cells in order to enhance their specificity and surpass immunosuppression. In recent years, many clinical studies have been conducted that eventually led to approved products. To date, the FDA has approved two anti-CD19 CAR T cell products for non-Hodgkin lymphoma therapy, i.e., axicbtagene ciloleucel and tisagenlecleucel. With all the advances that have been made in the field of CAR T cell therapy for hematologic malignancies therapy, ongoing studies are focused on optimizing its efficacy and specificity, as well as reducing the side effects. Also, the efforts are poised to broaden CAR T cell therapeutics for other cancers, especially solid tumors.
Collapse
Affiliation(s)
- Maryam Akhoundi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahsa Mohammadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Seyedeh Saeideh Sahraei
- Department of Reproductive Biology, Academic Center for Education, Culture and Research, Qom Branch, Qom, Iran.,Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom Branch, Qom, Iran
| | - Mohsen Sheykhhasan
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran. .,Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom Branch, Qom, Iran.
| | - Nashmin Fayazi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
27
|
Autologous antigen-presenting cells efficiently expand piggyBac transposon CAR-T cells with predominant memory phenotype. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:315-324. [PMID: 33898630 PMCID: PMC8047430 DOI: 10.1016/j.omtm.2021.03.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/17/2021] [Indexed: 12/26/2022]
Abstract
The quality of chimeric antigen receptor (CAR)-T cell products, including the expression of memory and exhaustion markers, has been shown to influence their long-term functionality. The manufacturing process of CAR-T cells should be optimized to prevent early T cell exhaustion during expansion. Activation of T cells by monoclonal antibodies is a critical step for T cell expansion, which may sometimes induce excess stimulation and exhaustion of T cells. Given that piggyBac transposon (PB)-based gene transfer could circumvent the conventional pre-activation of T cells, we established a manufacturing method of PB-mediated HER2-specific CAR-T cells (PB-HER2-CAR-T cells) that maintains their memory phenotype without early T cell exhaustion. Through stimulation of CAR-transduced T cells with autologous peripheral blood mononuclear cell-derived feeder cells expressing both truncated HER2, CD80, and 4-1BBL proteins, we could effectively propagate memory-rich, PD-1-negative PB-HER2-CAR-T cells. PB-HER2-CAR-T cells demonstrated sustained antitumor efficacy in vitro and debulked the HER2-positive tumors in vivo. Mice treated with PB-HER2-CAR-T cells rejected the second tumor establishment owing to the in vivo expansion of PB-HER2-CAR-T cells. Our simple and effective manufacturing process using PB system and genetically modified donor-derived feeder cells is a promising strategy for the use of PB-CAR-T cell therapy.
Collapse
|
28
|
Zhang Y, Wang P, Wang T, Fang Y, Ding Y, Qian Q. Chimeric antigen receptor T cells engineered to secrete CD40 agonist antibodies enhance antitumor efficacy. J Transl Med 2021; 19:82. [PMID: 33602263 PMCID: PMC7890961 DOI: 10.1186/s12967-021-02750-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/11/2021] [Indexed: 02/07/2023] Open
Abstract
Background Although chimeric antigen receptor (CAR)-T cell therapy has been remarkably successful for haematological malignancies, its efficacy against solid tumors is limited. The combination of CAR-T cell therapy with immune checkpoint inhibitors (CPIs), such as PD-1, PD-L1, and CTLA-4 antibodies, is a promising strategy for enhancing the antitumor efficacy of CAR-T cells. However, because most patients acquire resistance to CPIs, investigating other strategies is necessary to further improve the antitumor efficacy of CAR-T cell therapy for solid tumors. Recently, CD40 agonist antibodies showed potential antitumor efficacy by activating the CD40 pathway. Results Based on the piggyBac transposon system, rather than the widely used viral vectors, we constructed a meso3-CD40 CAR-T targeting region III of mesothelin (MSLN) that possessed the ability to secrete anti-CD40 antibodies. Compared with meso3 CAR-T cells, which did not secrete the anti-CD40 antibody, meso3-CD40 CAR-T cells secreted more cytokines and had a relatively higher proportion of central memory T (TCM) cells after stimulation by the target antigen. In addition, compared with meso3 CAR-T cells, meso3-CD40 CAR-T cells had a more powerful cytotoxic effect on target cells at a relatively low effector-to-target ratio. More importantly, we demonstrated that the antitumor activity of meso3-CD40 CAR-T cells was enhanced in a human ovarian cancer xenograft model in vivo. Conclusions In conclusion, these results highlight anti-CD40-secreting CAR-T cells generated by nonviral vectors as a potential clinical strategy for improving the efficacy of CAR-T cell therapies.
Collapse
Affiliation(s)
- Yajun Zhang
- Department of Biotherapy, Eastern Hepatobiliary Surgery Hospital, Navy Medical University, Shanghai, 201805, China
| | - Pei Wang
- Shanghai Engineering Research Center for Cell Therapy, Shanghai, 201805, China
| | - Tengjiao Wang
- Department of Bioinformatics, Institute of Translational Medicine, Navy Medical University, Shanghai, 201805, China
| | - Yuan Fang
- Shanghai Engineering Research Center for Cell Therapy, Shanghai, 201805, China.,College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, Zhejiang, China
| | - Yongmei Ding
- Department of Biotherapy, Eastern Hepatobiliary Surgery Hospital, Navy Medical University, Shanghai, 201805, China
| | - Qijun Qian
- Department of Biotherapy, Eastern Hepatobiliary Surgery Hospital, Navy Medical University, Shanghai, 201805, China. .,Shanghai Engineering Research Center for Cell Therapy, Shanghai, 201805, China. .,Department of Medical Oncology, Shanghai Mengchao Cancer Hospital, Shanghai, 201805, China. .,Shanghai University Cell Therapy Innovation Research Institute, Shanghai, 201805, China.
| |
Collapse
|
29
|
Abstract
Multiple myeloma remains an incurable disease despite great advances in its therapeutic landscape. Increasing evidence supports the belief that immune dysfunction plays an important role in the disease pathogenesis, progression, and drug resistance. Recent efforts have focused on harnessing the immune system to exert anti-myeloma effects with encouraging outcomes. First-in-class anti-CD38 monoclonal antibody, daratumumab, now forms part of standard treatment regimens in relapsed and refractory settings and is shifting to front-line treatments. However, a non-negligible number of patients will progress and be triple refractory from the first line of treatment. Antibody-drug conjugates, bispecific antibodies, and chimeric antigen receptors (CAR) are being developed in a heavily pretreated setting with outstanding results. Belantamab mafodotin-blmf has already received approval and other anti-B-cell maturation antigen (BCMA) therapies (CARs and bispecific antibodies are expected to be integrated in therapeutic options against myeloma soon. Nonetheless, immunotherapy faces different challenges in terms of efficacy and safety, and manufacturing and economic drawbacks associated with such a line of therapy pose additional obstacles to broadening its use. In this review, we described the most important clinical data on immunotherapeutic agents, delineated the limitations that lie in immunotherapy, and provided potential insights to overcome such issues.
Collapse
|
30
|
Angelis A, Naci H, Hackshaw A. Recalibrating Health Technology Assessment Methods for Cell and Gene Therapies. PHARMACOECONOMICS 2020; 38:1297-1308. [PMID: 32960434 DOI: 10.1007/s40273-020-00956-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Recently licensed cell and gene therapies have promising but highly uncertain clinical benefits. They are entering the market at very high prices, with the latest entrants costing hundreds of thousands of dollars. The significant long-term uncertainty posed by these therapies has already complicated the use of conventional economic evaluation approaches such as cost-effectiveness and cost-utility analyses, which are widely used for assessing the value of new health interventions. Cell and gene therapies also risk jeopardising healthcare systems' financial sustainability. As a result, there is a need to recalibrate the current health technology assessment methods used to measure and compensate their value. In this paper, we outline a set of technical adaptations and methodological refinements to address key challenges in the appraisal of cell and gene therapies' value, including the assessment of efficiency and affordability. We also discuss the potential role of alternative financing mechanisms. Ultimately, uncertainties associated with cell and gene therapies can only be meaningfully addressed by improving the evidence base supporting their approval and adoption in healthcare systems.
Collapse
Affiliation(s)
- Aris Angelis
- Department of Health Policy, London School of Economics and Political Science, Cowdray House, Portugal Street, London, UK.
| | - Huseyin Naci
- Department of Health Policy, London School of Economics and Political Science, Cowdray House, Portugal Street, London, UK
| | - Allan Hackshaw
- Cancer Research UK and UCL Cancer Trials Centre, UCL Cancer Institute, University College London, London, UK
| |
Collapse
|
31
|
Yang LR, Li L, Meng MY, Wang WJ, Yang SL, Zhao YY, Wang RQ, Gao H, Tang WW, Yang Y, Yang LL, Liao LW, Hou ZL. Evaluation of piggyBac-mediated anti-CD19 CAR-T cells after ex vivo expansion with aAPCs or magnetic beads. J Cell Mol Med 2020; 25:686-700. [PMID: 33225580 PMCID: PMC7812273 DOI: 10.1111/jcmm.16118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/30/2020] [Accepted: 11/04/2020] [Indexed: 12/02/2022] Open
Abstract
Adoptive immunotherapy is a new potential method of tumour therapy, among which anti‐CD19 chimeric antigen receptor T‐cell therapy (CAR‐T cell), is a typical treatment agent for haematological malignancies. Previous clinical trials showed that the quality and phenotype of CAR‐T cells expanded ex vivo would seriously affect the tumour treatment efficacy. Although magnetic beads are currently widely used to expand CAR‐T cells, the optimal expansion steps and methods have not been completely established. In this study, the differences between CAR‐T cells expanded with anti‐CD3/CD28 mAb‐coated beads and those expanded with cell‐based aAPCs expressing CD19/CD64/CD86/CD137L/mIL‐15 counter‐receptors were compared. The results showed that the number of CD19‐specific CAR‐T cells with a 4‐1BB and CD28 co‐stimulatory domain was much greater with stimulation by aAPCs than that with beads. In addition, the expression of memory marker CD45RO was higher, whereas expression of exhausted molecules was lower in CAR‐T cells expanded with aAPCs comparing with the beads. Both CAR‐T cells showed significant targeted tumoricidal effects. The CAR‐T cells stimulated with aAPCs secreted apoptosis‐related cytokines. Moreover, they also possessed marked anti‐tumour effect on NAMALWA xenograft mouse model. The present findings provided evidence on the safety and advantage of two expansion methods for CAR‐T cells genetically modified by piggyBac transposon system.
Collapse
Affiliation(s)
- Li-Rong Yang
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China.,Kunming Medical University, Kunming, China
| | - Lin Li
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, China
| | - Ming-Yao Meng
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, China
| | - Wen-Ju Wang
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, China
| | - Song-Lin Yang
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China.,Kunming Medical University, Kunming, China
| | - Yi-Yi Zhao
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, China
| | - Run-Qing Wang
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China.,Kunming Medical University, Kunming, China
| | - Hui Gao
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, China
| | - Wei-Wei Tang
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, China
| | - Yang Yang
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China.,Kunming Medical University, Kunming, China
| | - Li-Li Yang
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China.,Kunming Medical University, Kunming, China
| | - Li-Wei Liao
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, China
| | - Zong-Liu Hou
- Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, China
| |
Collapse
|
32
|
Lanitis E, Rota G, Kosti P, Ronet C, Spill A, Seijo B, Romero P, Dangaj D, Coukos G, Irving M. Optimized gene engineering of murine CAR-T cells reveals the beneficial effects of IL-15 coexpression. J Exp Med 2020; 218:211522. [PMID: 33156338 PMCID: PMC7653685 DOI: 10.1084/jem.20192203] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 07/24/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022] Open
Abstract
Limited clinical benefit has been demonstrated for chimeric antigen receptor (CAR) therapy of solid tumors, but coengineering strategies to generate so-called fourth-generation (4G) CAR-T cells are advancing toward overcoming barriers in the tumor microenvironment (TME) for improved responses. In large part due to technical challenges, there are relatively few preclinical CAR therapy studies in immunocompetent, syngeneic tumor-bearing mice. Here, we describe optimized methods for the efficient retroviral transduction and expansion of murine T lymphocytes of a predominantly central memory T cell (TCM cell) phenotype. We present a bicistronic retroviral vector encoding both a tumor vasculature–targeted CAR and murine interleukin-15 (mIL-15), conferring enhanced effector functions, engraftment, tumor control, and TME reprogramming, including NK cell activation and reduced presence of M2 macrophages. The 4G-CAR-T cells coexpressing mIL-15 were further characterized by up-regulation of the antiapoptotic marker Bcl-2 and lower cell-surface expression of the inhibitory receptor PD-1. Overall, this work introduces robust tools for the development and evaluation of 4G-CAR-T cells in immunocompetent mice, an important step toward the acceleration of effective therapies reaching the clinic.
Collapse
Affiliation(s)
- Evripidis Lanitis
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Giorgia Rota
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Paris Kosti
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Catherine Ronet
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | - Bili Seijo
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Pedro Romero
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Denarda Dangaj
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Melita Irving
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
33
|
Lee KH, Gowrishankar K, Street J, McGuire HM, Luciani F, Hughes B, Singh M, Clancy LE, Gottlieb DJ, Micklethwaite KP, Blyth E. Ex vivo enrichment of PRAME antigen-specific T cells for adoptive immunotherapy using CD137 activation marker selection. Clin Transl Immunology 2020; 9:e1200. [PMID: 33101678 PMCID: PMC7577233 DOI: 10.1002/cti2.1200] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/22/2020] [Accepted: 09/29/2020] [Indexed: 12/28/2022] Open
Abstract
Objective Adoptive immunotherapy with ex vivo expanded tumor‐specific T cells has potential as anticancer therapy. Preferentially expressed antigen in melanoma (PRAME) is an attractive target overexpressed in several cancers including melanoma and acute myeloid leukaemia (AML), with low expression in normal tissue outside the gonads. We developed a GMP‐compliant manufacturing method for PRAME‐specific T cells from healthy donors for adoptive immunotherapy. Methods Mononuclear cells were pulsed with PRAME 15‐mer overlapping peptide mix. After 16 h, activated cells expressing CD137 were isolated with immunomagnetic beads and cocultured with irradiated CD137neg fraction in medium supplemented with interleukin (IL)‐2, IL‐7 and IL‐15. Cultured T cells were restimulated with antigen‐pulsed autologous cells after 10 days. Cellular phenotype and cytokine response following antigen re‐exposure were assessed with flow cytometry, enzyme‐linked immunospot (ELISPOT) and supernatant cytokine detection. Detailed phenotypic and functional analysis with mass cytometry and T‐cell receptor (TCR) beta clonality studies were performed on selected cultures. Results PRAME‐stimulated cultures (n = 10) had mean expansion of 2500‐fold at day 18. Mean CD3+ percentage was 96% with CD4:CD8 ratio of 4:1. Re‐exposure to PRAME peptide mixture showed enrichment of CD4 cells expressing interferon (IFN)‐γ (mean: 12.2%) and TNF‐α (mean: 19.7%). Central and effector memory cells were 23% and 72%, respectively, with 24% T cells expressing PD1. Mass cytometry showed predominance of Th1 phenotype (CXCR3+/CCR4neg/CCR6neg/Tbet+, mean: 73%) and cytokine production including IL‐2, IL‐4, IL‐8, IL‐13 and GM‐CSF (2%, 6%, 8%, 4% and 11%, respectively). Conclusion PRAME‐specific T cells for adoptive immunotherapy were enriched from healthy donor mononuclear cells. The products were oligoclonal, exhibited Th1 phenotype and produced multiple cytokines.
Collapse
Affiliation(s)
- Koon H Lee
- Westmead Institute for Medical Research Westmead NSW Australia.,Faculty of Medicine and Health Sydney Medical School Sydney NSW Australia
| | | | - Janine Street
- Westmead Institute for Medical Research Westmead NSW Australia
| | - Helen M McGuire
- Faculty of Medicine and Health Sydney Medical School Sydney NSW Australia.,Ramaciotti Facility for Human Systems Biology The University of Sydney Sydney NSW Australia.,Charles Perkins Centre University of Sydney Sydney NSW Australia.,Discipline of Pathology Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia
| | - Fabio Luciani
- The Kirby Institute University of New South Wales Darlinghurst NSW Australia
| | - Brendan Hughes
- The Kirby Institute University of New South Wales Darlinghurst NSW Australia
| | - Mandeep Singh
- The Garvan Institute of Medical Research Darlinghurst NSW Australia.,St Vincent's Clinical School Faculty of Medicine UNSW Sydney Sydney NSW Australia
| | - Leighton E Clancy
- Westmead Institute for Medical Research Westmead NSW Australia.,Sydney Cellular Therapies Laboratory Westmead NSW Australia
| | - David J Gottlieb
- Westmead Institute for Medical Research Westmead NSW Australia.,Faculty of Medicine and Health Sydney Medical School Sydney NSW Australia.,Department of Haematology Westmead Hospital Westmead NSW Australia
| | - Kenneth P Micklethwaite
- Westmead Institute for Medical Research Westmead NSW Australia.,Faculty of Medicine and Health Sydney Medical School Sydney NSW Australia.,Department of Haematology Westmead Hospital Westmead NSW Australia
| | - Emily Blyth
- Westmead Institute for Medical Research Westmead NSW Australia.,Faculty of Medicine and Health Sydney Medical School Sydney NSW Australia.,Department of Haematology Westmead Hospital Westmead NSW Australia
| |
Collapse
|
34
|
Fritsche E, Volk HD, Reinke P, Abou-El-Enein M. Toward an Optimized Process for Clinical Manufacturing of CAR-Treg Cell Therapy. Trends Biotechnol 2020; 38:1099-1112. [DOI: 10.1016/j.tibtech.2019.12.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/21/2019] [Accepted: 12/06/2019] [Indexed: 02/08/2023]
|
35
|
Štach M, Ptáčková P, Mucha M, Musil J, Klener P, Otáhal P. Inducible secretion of IL-21 augments anti-tumor activity of piggyBac-manufactured chimeric antigen receptor T cells. Cytotherapy 2020; 22:744-754. [PMID: 32950390 DOI: 10.1016/j.jcyt.2020.08.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 08/11/2020] [Accepted: 08/13/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND The efficiency of chimeric antigen receptor (CAR) T-cell-based therapies depends on a sufficient expansion of CAR T cells in vivo and can be weakened by intra-tumoral suppression of CAR T cell functions, leading to a failure of therapy. For example, certain B-cell malignancies such as chronic lymphocytic leukemia are weakly sensitive to treatment with CAR T cells. Co-expression of proinflamatory cytokines such as IL-12 and IL-18 by CAR T cells have been shown to enhance their antitumor function. We similarly engineered CAR T cell to co-express IL-21 and studied the effects of IL-21 on CAR T cells specific to CD19 and prostate-specific membrane antigens using an in vitro co-culture model and NSG mice transplanted with B-cell tumors. RESULTS IL-21 enhanced the expansion of CAR T cells after antigenic stimulation, reduced the level of apoptosis of CAR T cells during co-culture with tumor cells and prevented differentiation of CAR T cells toward late memory phenotypes. In addition, induced secretion of IL-21 by CAR T cells promoted tumor infiltration by CD19-specific CAR (CAR19) T cells in NSG mice, resulting in reduced tumor growth. By co-culturing CAR19 T cells with bone-marrow fragments infiltrated with CLL cells we demonstrate that IL-21 reduces the immunosupressive activity of CLL cells against CAR19 T cells. CONCLUSIONS CAR19 T cells armed with IL-21 exhibited enhanced antitumor functions. IL-21 promoted their proliferation and cytotoxicity against chronic lymphocytic leukemia (CLL). The results suggest that arming CAR T cells with IL-21 could boost the effectiveness of CAR T-mediated therapies.
Collapse
Affiliation(s)
- Martin Štach
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Faculty of Natural Sciences, Charles University, Prague, Czech Republic
| | - Pavlína Ptáčková
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic; First Department of Medicine, Department of Haematology, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| | - Martin Mucha
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Faculty of Natural Sciences, Charles University, Prague, Czech Republic
| | - Jan Musil
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Pavel Klener
- First Department of Medicine, Department of Haematology, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| | - Pavel Otáhal
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic; First Department of Medicine, Department of Haematology, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic.
| |
Collapse
|
36
|
Overhauling CAR T Cells to Improve Efficacy, Safety and Cost. Cancers (Basel) 2020; 12:cancers12092360. [PMID: 32825533 PMCID: PMC7564591 DOI: 10.3390/cancers12092360] [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: 07/06/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/13/2022] Open
Abstract
Gene therapy is now surpassing 30 years of clinical experience and in that time a variety of approaches has been applied for the treatment of a wide range of pathologies. While the promise of gene therapy was over-stated in the 1990’s, the following decades were met with polar extremes between demonstrable success and devastating setbacks. Currently, the field of gene therapy is enjoying the rewards of overcoming the hurdles that come with turning new ideas into safe and reliable treatments, including for cancer. Among these modalities, the modification of T cells with chimeric antigen receptors (CAR-T cells) has met with clear success and holds great promise for the future treatment of cancer. We detail a series of considerations for the improvement of the CAR-T cell approach, including the design of the CAR, routes of gene transfer, introduction of CARs in natural killer and other cell types, combining the CAR approach with checkpoint blockade or oncolytic viruses, improving pre-clinical models as well as means for reducing cost and, thus, making this technology more widely available. While CAR-T cells serve as a prime example of translating novel ideas into effective treatments, certainly the lessons learned will serve to accelerate the current and future development of gene therapy drugs.
Collapse
|
37
|
Agarwalla P, Ogunnaike EA, Ahn S, Ligler FS, Dotti G, Brudno Y. Scaffold-Mediated Static Transduction of T Cells for CAR-T Cell Therapy. Adv Healthc Mater 2020; 9:e2000275. [PMID: 32592454 DOI: 10.1002/adhm.202000275] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/08/2020] [Indexed: 01/08/2023]
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has produced impressive clinical responses in patients with B-cell malignancies. Critical to the success of CAR-T cell therapies is the achievement of robust gene transfer into T cells mediated by viral vectors such as gamma-retroviral vectors. However, current methodologies of retroviral gene transfer rely on spinoculation and the use of retronectin, which may limit the implementation of cost-effective CAR-T cell therapies. Herein, a low-cost, tunable, macroporous, alginate scaffold that transduces T cells with retroviral vectors under static condition is described. CAR-T cells produced by macroporous scaffold-mediated viral transduction exhibit >60% CAR expression, retain effector phenotype, expand to clinically relevant cell numbers, and eradicate CD19+ lymphoma in vivo. Efficient transduction is dependent on scaffold macroporosity. Taken together, the data show that macroporous alginate scaffolds serve as an attractive alternative to current transduction protocols and have high potential for clinical translation to genetically modify T cells for adoptive cellular therapy.
Collapse
Affiliation(s)
- Pritha Agarwalla
- Joint Department of Biomedical Engineering, University of North Carolina ‐ Chapel Hill and North Carolina State University ‐ Raleigh 1840 Enterpreneur Way Raleigh NC 27695 USA
| | - Edikan A. Ogunnaike
- Department of Microbiology and ImmunologyUniversity of North Carolina at Chapel Hill Chapel Hill NC 27599 USA
| | - Sarah Ahn
- Department of Microbiology and ImmunologyUniversity of North Carolina at Chapel Hill Chapel Hill NC 27599 USA
| | - Frances S. Ligler
- Joint Department of Biomedical Engineering, University of North Carolina ‐ Chapel Hill and North Carolina State University ‐ Raleigh 1840 Enterpreneur Way Raleigh NC 27695 USA
| | - Gianpietro Dotti
- Department of Microbiology and ImmunologyUniversity of North Carolina at Chapel Hill Chapel Hill NC 27599 USA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina Chapel Hill. 450 West Dr. Chapel Hill NC 27599 USA
| | - Yevgeny Brudno
- Joint Department of Biomedical Engineering, University of North Carolina ‐ Chapel Hill and North Carolina State University ‐ Raleigh 1840 Enterpreneur Way Raleigh NC 27695 USA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina Chapel Hill. 450 West Dr. Chapel Hill NC 27599 USA
| |
Collapse
|
38
|
Fiorenza S, Ritchie DS, Ramsey SD, Turtle CJ, Roth JA. Value and affordability of CAR T-cell therapy in the United States. Bone Marrow Transplant 2020; 55:1706-1715. [PMID: 32474570 DOI: 10.1038/s41409-020-0956-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/13/2020] [Accepted: 05/18/2020] [Indexed: 12/22/2022]
Abstract
In the United States the increasing number of Food and Drug Administration (FDA)-approved, innovative, and potentially effective commercial cancer therapies pose a significant financial burden on public and private payers. Chimeric antigen receptor (CAR) T cells are prototypical of this challenge. In 2017 and 2018, tisagenlecleucel (Kymriah, Novartis) and axicabtagene ciloleucel (Yescarta, Kite) were approved by the FDA for use after showing groundbreaking results in relapsed/refractory B-cell malignancies. In 2020 and 2021, four further submissions to the FDA are expected for CAR T-cell therapies for indolent and aggressive B-cell malignancies and plasma cell myeloma. Yet, with marketed prices of over $350,000 per infusion for the two FDA-approved therapies and similar price tags expected for the coming products, serious concerns are raised over value and affordability. In this review we summarize recent, peer-reviewed cost-effectiveness studies of tisagenlecleucel and axicabtagene ciloleucel in the United States; discuss key issues concerning the health plan budget impact of CAR T-cell therapy; and review policy, payment and scientific approaches that may improve the value and affordability of CAR T-cell therapy.
Collapse
Affiliation(s)
- Salvatore Fiorenza
- Clinical Research Division and Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - David S Ritchie
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia.,Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Scott D Ramsey
- Hutchinson Institute for Cancer Outcomes Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Comparative Health Outcomes, Policy and Economics Institute, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - Cameron J Turtle
- Clinical Research Division and Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Medicine, University of Washington, Seattle, WA, USA
| | - Joshua A Roth
- Hutchinson Institute for Cancer Outcomes Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA. .,Comparative Health Outcomes, Policy and Economics Institute, School of Pharmacy, University of Washington, Seattle, WA, USA.
| |
Collapse
|
39
|
Magnani CF, Tettamanti S, Alberti G, Pisani I, Biondi A, Serafini M, Gaipa G. Transposon-Based CAR T Cells in Acute Leukemias: Where are We Going? Cells 2020; 9:cells9061337. [PMID: 32471151 PMCID: PMC7349235 DOI: 10.3390/cells9061337] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/21/2020] [Accepted: 05/25/2020] [Indexed: 02/07/2023] Open
Abstract
Chimeric Antigen Receptor (CAR) T-cell therapy has become a new therapeutic reality for refractory and relapsed leukemia patients and is also emerging as a potential therapeutic option in solid tumors. Viral vector-based CAR T-cells initially drove these successful efforts; however, high costs and cumbersome manufacturing processes have limited the widespread clinical implementation of CAR T-cell therapy. Here we will discuss the state of the art of the transposon-based gene transfer and its application in CAR T immunotherapy, specifically focusing on the Sleeping Beauty (SB) transposon system, as a valid cost-effective and safe option as compared to the viral vector-based systems. A general overview of SB transposon system applications will be provided, with an update of major developments, current clinical trials achievements and future perspectives exploiting SB for CAR T-cell engineering. After the first clinical successes achieved in the context of B-cell neoplasms, we are now facing a new era and it is paramount to advance gene transfer technology to fully exploit the potential of CAR T-cells towards next-generation immunotherapy.
Collapse
|
40
|
CAR T Cell Generation by piggyBac Transposition from Linear Doggybone DNA Vectors Requires Transposon DNA-Flanking Regions. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:359-368. [PMID: 32071928 PMCID: PMC7016334 DOI: 10.1016/j.omtm.2019.12.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 12/21/2019] [Indexed: 01/27/2023]
Abstract
CD19-specific chimeric antigen receptor (CAR19) T cells, generated using viral vectors, are an efficacious but costly treatment for B cell malignancies. The nonviral piggyBac transposon system provides a simple and inexpensive alternative for CAR19 T cell production. Until now, piggyBac has been plasmid based, facilitating economical vector amplification in bacteria. However, amplified plasmids have several undesirable qualities for clinical translation, including bacterial genetic elements, antibiotic-resistance genes, and the requirement for purification to remove endotoxin. Doggybones (dbDNA) are linear, covalently closed, minimal DNA vectors that can be inexpensively produced enzymatically in vitro at large scale. Importantly, they lack the undesirable features of plasmids. We used dbDNA incorporating piggyBac to generate CAR19 T cells. Initially, expression of functional transposase was evident, but stable CAR expression did not occur. After excluding other causes, additional random DNA flanking the transposon within the dbDNA was introduced, promoting stable CAR expression comparable to that of using plasmid components. Our findings demonstrate that dbDNA incorporating piggyBac can be used to generate CAR T cells and indicate that there is a requirement for DNA flanking the piggyBac transposon to enable effective transposition. dbDNA may further reduce the cost and improve the safety of CAR T cell production with transposon systems.
Collapse
|
41
|
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has revolutionized the immunotherapy field with high rate complete responses especially for hematological diseases. Despite the diversity of tumor specific-antigens, the manufacturing process is consistent and involves multiple steps, including selection of T cells, activation, genetic modification, and in vitro expansion. Among these complex manufacturing phases, the choice of culture system to generate a high number of functional cells needs to be evaluated and optimized. Flasks, bags, and rocking motion bioreactor are the most used platforms for CAR-T cell expansion in the current clinical trials but are far from being standardized. New processing options are available and a systematic effort seeking automation, standardization and the increase of production scale, would certainly help to bring the costs down and ultimately democratize this personalized therapy. In this review, we describe different cell expansion platforms available as well as the quality control requirements for clinical-grade production.
Collapse
Affiliation(s)
- Amanda Mizukami
- Center for Cell-Based Therapy CTC, Regional Blood Center of Ribeirão Preto, University of São Paulo, São Paulo, Brazil.
| | - Kamilla Swiech
- Center for Cell-Based Therapy CTC, Regional Blood Center of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
42
|
McGowan E, Lin Q, Ma G, Yin H, Chen S, Lin Y. PD-1 disrupted CAR-T cells in the treatment of solid tumors: Promises and challenges. Biomed Pharmacother 2020; 121:109625. [PMID: 31733578 DOI: 10.1016/j.biopha.2019.109625] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 10/27/2019] [Accepted: 10/31/2019] [Indexed: 12/13/2022] Open
Abstract
Unprecedented efficacy of chimeric antigen receptor (CAR) T cell therapy in the treatment of hematologic malignancies brings new hope for patients with many cancer types including solid tumors. However, the challenges for CAR-T cell therapy in eradicating solid tumors are immense. To overcome these seemingly intractable hurdles, more "powerful" CAR-T cells with enhanced antitumor efficacy are required. Emerging data support that the anti-tumor activity of CAR-T cells can be enhanced significantly without evident toxicity through simultaneous PD-1 disruption by genome editing. This review focuses on the current progress of PD-1 gene disrupted CAR-T cells in cancer therapy. Here we discuss key rationales for this new combination strategy and summarize the available pre-clinical studies. An update is provided on human clinical studies and available registered cancer clinical trials using CAR-T cells with PD-1 disruption. Future prospects and challenges are also discussed.
Collapse
Affiliation(s)
- Eileen McGowan
- Central Laboratory, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - Qimou Lin
- Department of Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Guocai Ma
- Department of Anesthesiology, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Haibin Yin
- Guangzhou Anjie Biomedical Technology Co. Ltd, Guangzhou, China
| | - Size Chen
- Central Laboratory, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Treatment, Guangzhou, China
| | - Yiguang Lin
- Central Laboratory, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; School of Life Sciences, University of Technology Sydney, Sydney, Australia.
| |
Collapse
|
43
|
Gershovich PM, Karabelskii AV, Ulitin AB, Ivanov RA. The Role of Checkpoint Inhibitors and Cytokines in Adoptive Cell-Based Cancer Immunotherapy with Genetically Modified T Cells. BIOCHEMISTRY (MOSCOW) 2019; 84:695-710. [PMID: 31509722 DOI: 10.1134/s0006297919070022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This review focuses on the structure and molecular action mechanisms of chimeric antigen receptors (CARs) and major aspects of the manufacturing and clinical application of products for the CAR-T (CAR-modified T lymphocyte) therapy of hematological and solid tumors with special emphasis on the strategies for combined use of CAR-T therapy with immuno-oncological monoclonal antibodies (checkpoint inhibitors) and cytokines to boost survival, persistence, and antitumor efficacy of CAR-T therapy. The review also summarizes preclinical and clinical data on the additive effects of the combined use of CAR-T therapy with interleukins and monoclonal antibodies targeting immune checkpoints.
Collapse
Affiliation(s)
- P M Gershovich
- CJSC Biocad, St. Petersburg, 198515, Russia. .,St. Petersburg State Chemical Pharmaceutical Academy, St. Petersburg, 197376, Russia
| | - A V Karabelskii
- CJSC Biocad, St. Petersburg, 198515, Russia.,St. Petersburg State Chemical Pharmaceutical Academy, St. Petersburg, 197376, Russia
| | - A B Ulitin
- CJSC Biocad, St. Petersburg, 198515, Russia
| | - R A Ivanov
- CJSC Biocad, St. Petersburg, 198515, Russia
| |
Collapse
|
44
|
Han J, Gao F, Geng S, Ye X, Wang T, Du P, Cai Z, Fu Z, Zhao Z, Shi L, Li Q, Cai J. Minicircle DNA-Engineered CAR T Cells Suppressed Tumor Growth in Mice. Mol Cancer Ther 2019; 19:178-186. [PMID: 31582530 DOI: 10.1158/1535-7163.mct-19-0204] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 07/04/2019] [Accepted: 09/27/2019] [Indexed: 11/16/2022]
Abstract
Viral-based chimeric antigen receptor-engineered T (CAR T)-cell manufacturing has potential safety risks and relatively high costs. The nonviral minicircle DNA (mcDNA) is safer for patients, cheaper to produce, and may be a more suitable technique to generate CAR T cells. In this study, we produced mcDNA-based CAR T cells specifically targeting prostate stem cell antigen (PSCA; mcDNA-PSCA-CAR T cells). Our results showed that mcDNA-PSCA-CAR T cells persisted in mouse peripheral blood as long as 28 days and demonstrated more CAR T-cell infiltration, higher cytokine secretion levels, and better antitumor effects. Together, our results suggest that mcDNA-CAR can be a safe and cost-effective platform to produce CAR T cells.
Collapse
Affiliation(s)
- Jinsheng Han
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fei Gao
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei, China.,Department of Surgery and Oncology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Songsong Geng
- Hebei Engineering Technology Research Center for Cell Therapy, Hebei HOFOY Biotech Company Ltd., Shijiazhuang, Hebei, China
| | - Xueshuai Ye
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei, China.,Hebei Engineering Technology Research Center for Cell Therapy, Hebei HOFOY Biotech Company Ltd., Shijiazhuang, Hebei, China
| | - Tie Wang
- Department of Surgery, Hebei Cangzhou Hospital of Integrated Traditional Chinese and Western Medicine, Cangzhou, Hebei, China
| | - Pingping Du
- Hebei Engineering Technology Research Center for Cell Therapy, Hebei HOFOY Biotech Company Ltd., Shijiazhuang, Hebei, China
| | - Ziqi Cai
- Hebei Engineering Technology Research Center for Cell Therapy, Hebei HOFOY Biotech Company Ltd., Shijiazhuang, Hebei, China
| | - Zexian Fu
- Hebei Engineering Technology Research Center for Cell Therapy, Hebei HOFOY Biotech Company Ltd., Shijiazhuang, Hebei, China.,Department of Surgery, Affiliated Hospital of Hebei Engineering University, Handan, Hebei, China
| | - Zhilong Zhao
- Hebei Engineering Technology Research Center for Cell Therapy, Hebei HOFOY Biotech Company Ltd., Shijiazhuang, Hebei, China.,Department of Surgery, the Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Long Shi
- Hebei Engineering Technology Research Center for Cell Therapy, Hebei HOFOY Biotech Company Ltd., Shijiazhuang, Hebei, China.,Department of Oncology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qingxia Li
- Department of Surgery and Oncology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Jianhui Cai
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei, China. .,Department of Surgery and Oncology, Hebei General Hospital, Shijiazhuang, Hebei, China.,Hebei Engineering Technology Research Center for Cell Therapy, Hebei HOFOY Biotech Company Ltd., Shijiazhuang, Hebei, China
| |
Collapse
|
45
|
Modified CAR T cells targeting membrane-proximal epitope of mesothelin enhances the antitumor function against large solid tumor. Cell Death Dis 2019; 10:476. [PMID: 31209210 PMCID: PMC6572851 DOI: 10.1038/s41419-019-1711-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/21/2019] [Accepted: 05/23/2019] [Indexed: 12/13/2022]
Abstract
Mesothelin (MSLN) is an attractive antigen for chimeric antigen receptor (CAR) T therapy and the epitope selection within MSLN is essential. In this study, we constructed two types of CARs targeting either region I of MSLN (meso1 CAR, also known as a membrane-distal region) or region III of MSLN (meso3 CAR, also known as a membrane-proximal region) using a modified piggyBac transposon system. We reported that, compared with meso1 CAR T cells, meso3 CAR T cells express higher levels of CD107α upon activation and produce increased levels of interleukin-2, TNF-α, and IFN-γ against multiple MSLN-expressing cancer cells in vitro. In a real-time cell analyzer system and a three-dimensional spheroid cancer cell model, we also demonstrated that meso3 CAR T cells display an enhanced killing effect compared with that of meso1 CAR T cells. More importantly, in a gastric cancer NSG mice model, meso3 CAR T cells mediated stronger antitumor responses than meso1 CAR T cells did. We further identified that meso3 CAR T cells can effectively inhibit the growth of large ovarian tumors in vivo. Collectively, our study provides evidences that meso3 CAR T-cell therapy performs as a better immunotherapy than meso1 CAR T-cell therapy in treating MSLN-positive solid tumors.
Collapse
|
46
|
Clauss J, Obenaus M, Miskey C, Ivics Z, Izsvák Z, Uckert W, Bunse M. Efficient Non-Viral T-Cell Engineering by Sleeping Beauty Minicircles Diminishing DNA Toxicity and miRNAs Silencing the Endogenous T-Cell Receptors. Hum Gene Ther 2019; 29:569-584. [PMID: 29562762 DOI: 10.1089/hum.2017.136] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Transposon-based vectors have entered clinical trials as an alternative to viral vectors for genetic engineering of T cells. However, transposon vectors require DNA transfection into T cells, which were found to cause adverse effects. T-cell viability was decreased in a dose-dependent manner, and DNA-transfected T cells showed a delayed response upon T-cell receptor (TCR) stimulation with regard to blast formation, proliferation, and surface expression of CD25 and CD28. Gene expression analysis demonstrated a DNA-dependent induction of a type I interferon response and interferon-β upregulation. By combining Sleeping Beauty transposon minicircle vectors with SB100X transposase-encoding RNA, it was possible to reduce the amount of total DNA required, and stable expression of therapeutic TCRs was achieved in >50% of human T cells without enrichment. The TCR-engineered T cells mediated effective tumor cell killing and cytokine secretion upon antigen-specific stimulation. Additionally, the Sleeping Beauty transposon system was further improved by miRNAs silencing the endogenous TCR chains. These miRNAs increased the surface expression of the transgenic TCR, diminished mispairing with endogenous TCR chains, and enhanced antigen-specific T-cell functionality. This approach facilitates the rapid non-viral generation of highly functional, engineered T cells for immunotherapy.
Collapse
Affiliation(s)
- Julian Clauss
- 1 Max Delbrück Center for Molecular Medicine in the Helmholtz Association , Berlin, Germany
| | - Matthias Obenaus
- 1 Max Delbrück Center for Molecular Medicine in the Helmholtz Association , Berlin, Germany .,2 Charité Universitätsmedizin Berlin , Campus Virchow-Klinikum, Berlin, Germany
| | - Csaba Miskey
- 3 Division of Medical Biotechnology, Paul Ehrlich-Institut , Langen, Germany
| | - Zoltán Ivics
- 3 Division of Medical Biotechnology, Paul Ehrlich-Institut , Langen, Germany
| | - Zsuzsanna Izsvák
- 1 Max Delbrück Center for Molecular Medicine in the Helmholtz Association , Berlin, Germany .,4 Berlin Institute of Health , Berlin, Germany
| | - Wolfgang Uckert
- 1 Max Delbrück Center for Molecular Medicine in the Helmholtz Association , Berlin, Germany .,4 Berlin Institute of Health , Berlin, Germany .,5 Institute of Biology, Humboldt-Universität zu Berlin , Berlin, Germany
| | - Mario Bunse
- 1 Max Delbrück Center for Molecular Medicine in the Helmholtz Association , Berlin, Germany
| |
Collapse
|
47
|
Mardiana S, Lai J, House IG, Beavis PA, Darcy PK. Switching on the green light for chimeric antigen receptor T-cell therapy. Clin Transl Immunology 2019; 8:e1046. [PMID: 31073403 PMCID: PMC6500780 DOI: 10.1002/cti2.1046] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/13/2019] [Accepted: 03/14/2019] [Indexed: 12/18/2022] Open
Abstract
Adoptive cellular therapy involving genetic modification of T cells with chimeric antigen receptor (CAR) transgene offers a promising strategy to broaden the efficacy of this approach for the effective treatment of cancer. Although remarkable antitumor responses have been observed following CAR T‐cell therapy in a subset of B‐cell malignancies, this has yet to be extended in the context of solid cancers. A number of promising strategies involving reprogramming the tumor microenvironment, increasing the specificity and safety of gene‐modified T cells and harnessing the endogenous immune response have been tested in preclinical models that may have a significant impact in patients with solid cancers. This review will discuss these exciting new developments and the challenges that must be overcome to deliver a more sustained and potent therapeutic response.
Collapse
Affiliation(s)
- Sherly Mardiana
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Junyun Lai
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Imran Geoffrey House
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Paul Andrew Beavis
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Phillip Kevin Darcy
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia.,Department of Pathology University of Melbourne Parkville VIC Australia.,Department of Immunology Monash University Clayton VIC Australia
| |
Collapse
|
48
|
Elavia N, Panch SR, McManus A, Bikkani T, Szymanski J, Highfill SL, Jin P, Brudno J, Kochenderfer J, Stroncek DF. Effects of starting cellular material composition on chimeric antigen receptor T-cell expansion and characteristics. Transfusion 2019; 59:1755-1764. [PMID: 30973976 DOI: 10.1111/trf.15287] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/03/2019] [Accepted: 01/03/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND When manufacturing chimeric antigen receptor (CAR) T cells using anti-CD3/anti-CD28 beads, ex vivo T-cell expansion is dependent on the composition of leukocytes used in the manufacturing process. We investigated the effects of leukocyte composition on CAR T-cell expansion and characteristics using an alternative manufacturing method. METHODS Anti-B-cell maturation antigen and CD19-CAR T cells were manufactured using autologous peripheral blood mononuclear cell (PBMNC) concentrates. The PBMNCs were enriched for lymphocytes using density gradient separation, which were used for CAR T-cell culture initiation. T-cell expansion was stimulated with soluble anti-CD3 and interleukin-2. RESULTS Fifty-one CAR T-cell products were evaluated; 28 anti-B-cell maturation antigen (BCMA) CAR T cells produced for 24 patients and 27 CD19 CAR T cells produced for 24 patients. CAR T-cell expansion was reduced when greater quantities of monocytes were present in the post-density gradient separation PBMNCs. In addition, the ratio of CD4 to CD8 cells in the CAR T-cell products after 7 days of culture was dependent on the quantity of monocytes, RBCs, and neutrophils in the post-density gradient separation PBMNCs. Greater quantities of monocytes and RBCs were associated with a greater proportion of CD4+ cells and greater quantities of neutrophils were associated with a greater proportion of CD8+ cells. CONCLUSIONS The composition of leukocytes used to manufacture CAR T cells can affect cell expansion and the composition of CAR T-cell products. More uniform or complete lymphocyte enrichment of PBMNCs improves the consistency of final CAR T-cell products.
Collapse
Affiliation(s)
- Nasha Elavia
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland.,Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Sandhya R Panch
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland.,Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Andrew McManus
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland.,Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Thejaswi Bikkani
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland.,Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - James Szymanski
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland.,Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Steven L Highfill
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland.,Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Ping Jin
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland.,Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Jennifer Brudno
- Experimental Transplantation and Immunology Branch, National Cancer Institute and National Institutes of Health, Bethesda, Maryland
| | - James Kochenderfer
- Experimental Transplantation and Immunology Branch, National Cancer Institute and National Institutes of Health, Bethesda, Maryland
| | - David F Stroncek
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland.,Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, Maryland
| |
Collapse
|
49
|
Mochel JP, Ekker SC, Johannes CM, Jergens AE, Allenspach K, Bourgois-Mochel A, Knouse M, Benzekry S, Wierson W, LeBlanc AK, Kenderian SS. CAR T Cell Immunotherapy in Human and Veterinary Oncology: Changing the Odds Against Hematological Malignancies. AAPS JOURNAL 2019; 21:50. [PMID: 30963322 DOI: 10.1208/s12248-019-0322-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 03/17/2019] [Indexed: 01/14/2023]
Abstract
The advent of the genome editing era brings forth the promise of adoptive cell transfer using engineered chimeric antigen receptor (CAR) T cells for targeted cancer therapy. CAR T cell immunotherapy is probably one of the most encouraging developments for the treatment of hematological malignancies. In 2017, two CAR T cell therapies were approved by the US Food and Drug Administration: one for the treatment of pediatric acute lymphoblastic leukemia (ALL) and the other for adult patients with advanced lymphomas. However, despite significant progress in the area, CAR T cell therapy is still in its early days and faces significant challenges, including the complexity and costs associated with the technology. B cell lymphoma is the most common hematopoietic cancer in dogs, with an incidence approaching 0.1% and a total of 20-100 cases per 100,000 individuals. It is a widely accepted naturally occurring model for human non-Hodgkin's lymphoma. Current treatment is with combination chemotherapy protocols, which prolong life for less than a year in canines and are associated with severe dose-limiting side effects, such as gastrointestinal and bone marrow toxicity. To date, one canine study generated CAR T cells by transfection of mRNA for CAR domain expression. While this was shown to provide a transient anti-tumor activity, results were modest, indicating that stable, genomic integration of CAR modules is required in order to achieve lasting therapeutic benefit. This commentary summarizes the current state of knowledge on CAR T cell immunotherapy in human medicine and its potential applications in animal health, while discussing the potential of the canine model as a translational system for immuno-oncology research.
Collapse
Affiliation(s)
- Jonathan P Mochel
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, 50011, USA. .,Iowa State University College of Vet. Medicine, 2448 Lloyd, 1809 S Riverside Dr., Ames, Iowa, 50011-1250, USA.
| | - Stephen C Ekker
- Mayo Clinic Cancer Center Department of Biochemistry and Molecular Biology, Rochester, Minnesota, 55905, USA
| | - Chad M Johannes
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, Iowa, 50011, USA
| | - Albert E Jergens
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, Iowa, 50011, USA
| | - Karin Allenspach
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, Iowa, 50011, USA
| | - Agnes Bourgois-Mochel
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, Iowa, 50011, USA
| | - Michael Knouse
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, 50011, USA
| | - Sebastien Benzekry
- Team MONC, Institut National de Recherche en Informatique et en Automatique, Bordeaux, France
| | - Wesley Wierson
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, Iowa, 50011, USA
| | - Amy K LeBlanc
- Comparative Oncology Program, Center for Cancer Research National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Saad S Kenderian
- Department of Medicine, Mayo Clinic Division of Hematology, Rochester, Minnesota, 55905, USA.,Department of Immunology, Mayo Clinic, Rochester, Minnesota, 55905, USA
| |
Collapse
|
50
|
Chicaybam L, Abdo L, Carneiro M, Peixoto B, Viegas M, de Sousa P, Fornazin MC, Spago MC, Albertoni Laranjeira AB, de Campos-Lima PO, Nowill A, Barros LRC, Bonamino MH. CAR T Cells Generated UsingSleeping BeautyTransposon Vectors and Expanded with an EBV-Transformed Lymphoblastoid Cell Line Display Antitumor ActivityIn VitroandIn Vivo. Hum Gene Ther 2019; 30:511-522. [DOI: 10.1089/hum.2018.218] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Leonardo Chicaybam
- Molecular Carcinogenesis Program, National Cancer Institute (INCA), Rio de Janeiro, Brazil
- Vice-Presidency of Research and Biological Collections, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Luiza Abdo
- Molecular Carcinogenesis Program, National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Mayra Carneiro
- Molecular Carcinogenesis Program, National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Bárbara Peixoto
- Cell Biology Program, National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Mariana Viegas
- Molecular Carcinogenesis Program, National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Priscila de Sousa
- Molecular Carcinogenesis Program, National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Márcia C. Fornazin
- Integrated Center for Oncohematology Research in Infancy, Institute of Biology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Maria C. Spago
- Integrated Center for Oncohematology Research in Infancy, Institute of Biology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | | | - Pedro O. de Campos-Lima
- Institute of Molecular and Cellular Engineering, Boldrini Children's Center, Campinas, Sao Paulo, Brazil
- Functional and Molecular Biology Program, Institute of Biology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Alexandre Nowill
- Integrated Center for Oncohematology Research in Infancy, Institute of Biology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | | | - Martín H. Bonamino
- Molecular Carcinogenesis Program, National Cancer Institute (INCA), Rio de Janeiro, Brazil
- Vice-Presidency of Research and Biological Collections, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|