1
|
Application and Design of Switches Used in CAR. Cells 2022; 11:cells11121910. [PMID: 35741039 PMCID: PMC9221702 DOI: 10.3390/cells11121910] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 11/21/2022] Open
Abstract
Among the many oncology therapies, few have generated as much excitement as CAR-T. The success of CAR therapy would not have been possible without the many discoveries that preceded it, most notably, the Nobel Prize-winning breakthroughs in cellular immunity. However, despite the fact that CAR-T already offers not only hope for development, but measurable results in the treatment of hematological malignancies, CAR-T still cannot be safely applied to solid tumors. The reason for this is, among other things, the lack of tumor-specific antigens which, in therapy, threatens to cause a lethal attack of lymphocytes on healthy cells. In the case of hematological malignancies, dangerous complications such as cytokine release syndrome may occur. Scientists have responded to these clinical challenges with molecular switches. They make it possible to remotely control CAR lymphocytes after they have already been administered to the patient. Moreover, they offer many additional capabilities. For example, they can be used to switch CAR antigenic specificity, create logic gates, or produce local activation under heat or light. They can also be coupled with costimulatory domains, used for the regulation of interleukin secretion, or to prevent CAR exhaustion. More complex modifications will probably require a combination of reprogramming (iPSc) technology with genome editing (CRISPR) and allogenic (off the shelf) CAR-T production.
Collapse
|
2
|
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
|
3
|
Lin Z, Liu X, Liu T, Gao H, Wang S, Zhu X, Rong L, Cheng J, Cai Z, Xu F, Tan X, Lv L, Li Z, Sun Y, Qian Q. Evaluation of Nonviral piggyBac and lentiviral Vector in Functions of CD19chimeric Antigen Receptor T Cells and Their Antitumor Activity for CD19 + Tumor Cells. Front Immunol 2022; 12:802705. [PMID: 35082789 PMCID: PMC8784881 DOI: 10.3389/fimmu.2021.802705] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/14/2021] [Indexed: 11/13/2022] Open
Abstract
Nonviral transposon piggyBac (PB) and lentiviral (LV) vectors have been used to deliver chimeric antigen receptor (CAR) to T cells. To understand the differences in the effects of PB and LV on CAR T-cell functions, a CAR targeting CD19 was cloned into PB and LV vectors, and the resulting pbCAR and lvCAR were delivered to T cells to generate CD19pbCAR and CD19lvCAR T cells. Both CD19CAR T-cell types were strongly cytotoxic and secreted high IFN-γ levels when incubated with Raji cells. TNF-α increased in CD19pbCAR T cells, whereas IL-10 increased in CD19lvCAR T cells. CD19pbCAR and CD19lvCAR T cells showed similar strong anti-tumor activity in Raji cell-induced mouse models, slightly reducing mouse weight while enhancing mouse survival. High, but not low or moderate, concentrations of CD19pbCAR T cells significantly inhibited Raji cell-induced tumor growth in vivo. These CD19pbCAR T cells were distributed mostly in mesenteric lymph nodes, bone marrow of the femur, spleen, kidneys, and lungs, specifically accumulating at CD19-rich sites and CD19-positive tumors, with CAR copy number being increased on day 7. These results indicate that pbCAR has its specific activities and functions in pbCAR T cells, making it a valuable tool for CAR T-cell immunotherapy.
Collapse
MESH Headings
- Animals
- Antigens, CD19/genetics
- Antigens, CD19/immunology
- Antigens, CD19/metabolism
- Cell Line, Tumor
- Cells, Cultured
- Cytotoxicity, Immunologic/immunology
- DNA Transposable Elements/genetics
- DNA Transposable Elements/immunology
- Female
- Genetic Vectors/genetics
- Genetic Vectors/immunology
- Humans
- Immunotherapy, Adoptive/methods
- Lentivirus/genetics
- Lentivirus/immunology
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Neoplasms/immunology
- Neoplasms/pathology
- Neoplasms/therapy
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Burden/immunology
- Xenograft Model Antitumor Assays/methods
- Mice
Collapse
Affiliation(s)
- Zhicai Lin
- Medical, Cell Product and R&D Department, Center for Cell Pharmaceuticals, Shanghai Cell Therapy Group, Shanghai, China
| | - Xiangzhen Liu
- Medical, Cell Product and R&D Department, Center for Cell Pharmaceuticals, Shanghai Cell Therapy Group, Shanghai, China
| | - Tao Liu
- R&D Department, Nucleotide Center, Shanghai Cell Therapy Group, Shanghai, China
| | - Haixia Gao
- R&D Department, Nucleotide Center, Shanghai Cell Therapy Group, Shanghai, China
| | - Sitong Wang
- Medical, Cell Product and R&D Department, Center for Cell Pharmaceuticals, Shanghai Cell Therapy Group, Shanghai, China
| | - Xingli Zhu
- Medical, Cell Product and R&D Department, Center for Cell Pharmaceuticals, Shanghai Cell Therapy Group, Shanghai, China
| | - Lijie Rong
- Medical, Cell Product and R&D Department, Center for Cell Pharmaceuticals, Shanghai Cell Therapy Group, Shanghai, China
| | - Jingbo Cheng
- Medical, Cell Product and R&D Department, Center for Cell Pharmaceuticals, Shanghai Cell Therapy Group, Shanghai, China
| | - Zhigang Cai
- Medical, Cell Product and R&D Department, Center for Cell Pharmaceuticals, Shanghai Cell Therapy Group, Shanghai, China
| | - Fu Xu
- Medical, Cell Product and R&D Department, Center for Cell Pharmaceuticals, Shanghai Cell Therapy Group, Shanghai, China
| | - Xue Tan
- Medical, Cell Product and R&D Department, Center for Cell Pharmaceuticals, Shanghai Cell Therapy Group, Shanghai, China
| | - Linjie Lv
- Medical, Cell Product and R&D Department, Center for Cell Pharmaceuticals, Shanghai Cell Therapy Group, Shanghai, China
| | - Zhong Li
- Medical, Cell Product and R&D Department, Center for Cell Pharmaceuticals, Shanghai Cell Therapy Group, Shanghai, China
- Department of Immunotherapy, Shanghai Cell Therapy Research Institute, Shanghai, China
| | - Yan Sun
- Medical, Cell Product and R&D Department, Center for Cell Pharmaceuticals, Shanghai Cell Therapy Group, Shanghai, China
| | - Qijun Qian
- Medical, Cell Product and R&D Department, Center for Cell Pharmaceuticals, Shanghai Cell Therapy Group, Shanghai, China
- Department of Immunotherapy, Shanghai Cell Therapy Research Institute, Shanghai, China
- Shanghai Menchao Cancer Hospital, Shanghai University, Shanghai, China
| |
Collapse
|
4
|
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
|
5
|
Endogenous Transposase Source in Human Cells Mobilizes piggyBac Transposons. Mol Ther 2017; 24:851-4. [PMID: 27198853 DOI: 10.1038/mt.2016.76] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
6
|
Klose D, Woitok M, Niesen J, Beerli RR, Grawunder U, Fischer R, Barth S, Fendel R, Nachreiner T. Generation of an artificial human B cell line test system using Transpo-mAbTM technology to evaluate the therapeutic efficacy of novel antigen-specific fusion proteins. PLoS One 2017; 12:e0180305. [PMID: 28704435 PMCID: PMC5509223 DOI: 10.1371/journal.pone.0180305] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 06/13/2017] [Indexed: 12/21/2022] Open
Abstract
The antigen-specific targeting of autoreactive B cells via their unique B cell receptors (BCRs) is a novel and promising alternative to the systemic suppression of humoral immunity. We generated and characterized cytolytic fusion proteins based on an existing immunotoxin comprising tetanus toxoid fragment C (TTC) as the targeting component and the modified Pseudomonas aeruginosa exotoxin A (ETA') as the cytotoxic component. The immunotoxin was reconfigured to replace ETA' with either the granzyme B mutant R201K or MAPTau as human effector domains. The novel cytolytic fusion proteins were characterized with a recombinant human lymphocytic cell line developed using Transpo-mAb™ technology. Genes encoding a chimeric TTC-reactive immunoglobulin G were successfully integrated into the genome of the precursor B cell line REH so that the cells could present TTC-reactive BCRs on their surface. These cells were used to investigate the specific cytotoxicity of GrB(R201K)-TTC and TTC-MAPTau, revealing that the serpin proteinase inhibitor 9-resistant granzyme B R201K mutant induced apoptosis specifically in the lymphocytic cell line. Our data confirm that antigen-based fusion proteins containing granzyme B (R201K) are suitable candidates for the depletion of autoreactive B cells.
Collapse
Affiliation(s)
- Diana Klose
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
- Department of Experimental Medicine and Immunotherapy, Institute for Applied Medical Engineering, University Hospital RWTH Aachen, Aachen, Germany
| | - Mira Woitok
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
- Institute of Molecular Biotechnology (Biology VII), RWTH Aachen University, Aachen, Germany
| | - Judith Niesen
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| | | | | | - Rainer Fischer
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
- Institute of Molecular Biotechnology (Biology VII), RWTH Aachen University, Aachen, Germany
| | - Stefan Barth
- Department of Experimental Medicine and Immunotherapy, Institute for Applied Medical Engineering, University Hospital RWTH Aachen, Aachen, Germany
- South African Research Chair in Cancer Biotechnology, Institute of Infectious Disease and Molecular Medicine (IDM), Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Rolf Fendel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
- * E-mail:
| | - Thomas Nachreiner
- Department of Experimental Medicine and Immunotherapy, Institute for Applied Medical Engineering, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
7
|
Zhu X, Prasad S, Gaedicke S, Hettich M, Firat E, Niedermann G. Patient-derived glioblastoma stem cells are killed by CD133-specific CAR T cells but induce the T cell aging marker CD57. Oncotarget 2015; 6:171-84. [PMID: 25426558 PMCID: PMC4381586 DOI: 10.18632/oncotarget.2767] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 11/14/2014] [Indexed: 02/07/2023] Open
Abstract
The AC133 epitope of CD133 is a cancer stem cell (CSC) marker for many tumor entities, including the highly malignant glioblastoma multiforme (GBM). We have developed an AC133-specific chimeric antigen receptor (CAR) and show that AC133-CAR T cells kill AC133+ GBM stem cells (GBM-SCs) both in vitro and in an orthotopic tumor model in vivo. Direct contact with patient-derived GBM-SCs caused rapid upregulation of CD57 on the CAR T cells, a molecule known to mark terminally or near-terminally differentiated T cells. However, other changes associated with terminal T cell differentiation could not be readily detected. CD57 is also expressed on tumor cells of neural crest origin and has been preferentially found on highly aggressive, undifferentiated, multipotent CSC-like cells. We found that CD57 was upregulated on activated T cells only upon contact with CD57+ patient-derived GBM-SCs, but not with conventional CD57-negative glioma lines. However, CD57 was not downregulated on the GBM-SCs upon their differentiation, indicating that this molecule is not a bona fide CSC marker for GBM. Differentiated GBM cells still induced CD57 on CAR T cells and other activated T cells. Therefore, CD57 can apparently be upregulated on activated human T cells by mere contact with CD57+ target cells.
Collapse
Affiliation(s)
- Xuekai Zhu
- Department of Radiation Oncology, University Hospital Freiburg, Freiburg, Germany
| | - Shruthi Prasad
- Department of Radiation Oncology, University Hospital Freiburg, Freiburg, Germany. Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Simone Gaedicke
- Department of Radiation Oncology, University Hospital Freiburg, Freiburg, Germany
| | - Michael Hettich
- Department of Radiation Oncology, University Hospital Freiburg, Freiburg, Germany. Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Elke Firat
- Department of Radiation Oncology, University Hospital Freiburg, Freiburg, Germany
| | - Gabriele Niedermann
- Department of Radiation Oncology, University Hospital Freiburg, Freiburg, Germany. German Cancer Consortium (DKTK), Freiburg, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
8
|
Anti-leukemic potency of piggyBac-mediated CD19-specific T cells against refractory Philadelphia chromosome-positive acute lymphoblastic leukemia. Cytotherapy 2015; 16:1257-69. [PMID: 25108652 DOI: 10.1016/j.jcyt.2014.05.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 05/25/2014] [Accepted: 05/30/2014] [Indexed: 11/21/2022]
Abstract
BACKGROUND AIMS To develop a treatment option for Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph(+)ALL) resistant to tyrosine kinase inhibitors (TKIs), we evaluated the anti-leukemic activity of T cells non-virally engineered to express a CD19-specific chimeric antigen receptor (CAR). METHODS A CD19.CAR gene was delivered into mononuclear cells from 10 mL of blood of healthy donors through the use of piggyBac-transposons and the 4-D Nucleofector System. Nucleofected cells were stimulated with CD3/CD28 antibodies, magnetically selected for the CD19.CAR, and cultured in interleukin-15-containing serum-free medium with autologous feeder cells for 21 days. To evaluate their cytotoxic potency, we co-cultured CAR T cells with seven Ph(+)ALL cell lines including three TKI-resistant (T315I-mutated) lines at an effector-to-target ratio of 1:5 or lower without cytokines. RESULTS We obtained ∼1.3 × 10(8) CAR T cells (CD4(+), 25.4%; CD8(+), 71.3%), co-expressing CD45RA and CCR7 up to ∼80%. After 7-day co-culture, CAR T cells eradicated all tumor cells at the 1:5 and 1:10 ratios and substantially reduced tumor cell numbers at the 1:50 ratio. Kinetic analysis revealed up to 37-fold proliferation of CAR T cells during a 20-day culture period in the presence of tumor cells. On exposure to tumor cells, CAR T cells transiently and reproducibly upregulated the expression of transgene as well as tumor necrosis factor-related apoptosis-inducing ligand and interleukin-2. CONCLUSIONS We generated a clinically relevant number of CAR T cells from 10 mL of blood through the use of piggyBac-transposons, a 4D-Nulcleofector, and serum/xeno/tumor cell/virus-free culture system. CAR T cells exhibited marked cytotoxicity against Ph(+)ALL regardless of T315I mutation. PiggyBac-mediated CD19-specific T-cell therapy may provide an effective, inexpensive and safe option for drug-resistant Ph(+)ALL.
Collapse
|