1
|
Dan L, Kang-Zheng L. Optimizing viral transduction in immune cell therapy manufacturing: key process design considerations. J Transl Med 2025; 23:501. [PMID: 40316943 PMCID: PMC12046913 DOI: 10.1186/s12967-025-06524-0] [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: 12/31/2024] [Accepted: 04/21/2025] [Indexed: 05/04/2025] Open
Abstract
Immune cell therapies have revolutionized the treatment of cancers, autoimmune disorders, and infectious diseases. A critical step in their manufacturing is viral transduction, which enables the delivery of therapeutic genes into immune cells. However, the complexity of this process presents significant challenges for optimization and scalability. This review provides a comprehensive analysis of viral transduction process in immune cell therapy manufacturing, highlighting key design considerations to support the development of safe, effective, and scalable production methods. Additionally, it examines current technological challenges in immune cell transduction and explores future innovations poised to advance the field.
Collapse
Affiliation(s)
- Liu Dan
- Bioprocessing Technology Institute BTI, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Singapore, 138668, Singapore.
| | - Lee Kang-Zheng
- Bioprocessing Technology Institute BTI, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Singapore, 138668, Singapore
| |
Collapse
|
2
|
Moter A, Scharf S, Schäfer H, Bexte T, Wendel P, Donnadieu E, Hansmann M, Hartmann S, Ullrich E. Migration Dynamics of Human NK Cell Preparations in Microchannels and Their Invasion Into Patient-Derived Tissue. J Cell Mol Med 2025; 29:e70481. [PMID: 40159644 PMCID: PMC11955413 DOI: 10.1111/jcmm.70481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Natural killer (NK) cells are characterised by their ability to attack cancer cells without prior antigen stimulation. Additionally, clinical trials revealed great potential of NK cells expressing chimeric antigen receptors (CARs). Successful anti-tumour efficacy remains limited by migration and infiltration to the tumour site by NK cell preparations, which is linked to the scarcity in the knowledge of migration dynamics and invasion potential. Here, we applied a recently reported innovative microfluidic microchannel technology to gain insight into the intrinsic motility of NK cells. We assessed the baseline activated and proliferating NK cells in direct comparison with T cells and investigated their motility patterns in the presence of tumour cells. Additionally, we performed high-resolution 4D confocal imaging in patient-derived hyperplastic lymphatic tissues to assess their invasive capacity. Our data revealed that the invasion potential of NK cells was greater than that of T cells, despite their similar velocities. The flexibility of the NK cell nucleus may have contributed to the higher invasion potential. The motility of CD19-CAR-NK cell preparations was similar to that of non-transduced NK cells in hyperplastic lymphoid tissue, with improved targeted migration in tumour tissue, suggesting the suitability of genetically engineered NK cells for difficult-to-reach tumour tissues.
Collapse
Affiliation(s)
- Alina Moter
- Goethe University Frankfurt, Department of Pediatrics, Experimental Immunology and Cell TherapyFrankfurt (Main)Germany
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI)Frankfurt (Main)Germany
| | - Sonja Scharf
- Institute of PathologyUniversity Hospital Essen, University of Duisburg‐EssenEssenGermany
| | - Hendrik Schäfer
- Institute of PathologyUniversity Hospital Essen, University of Duisburg‐EssenEssenGermany
| | - Tobias Bexte
- Goethe University Frankfurt, Department of Pediatrics, Experimental Immunology and Cell TherapyFrankfurt (Main)Germany
- Institute for Transfusion Medicine and ImmunohematologyGerman Red Cross Blood Service Baden‐Württemberg – HessenHessenGermany
- Goethe University Frankfurt, University Cancer Center Frankfurt (UCT), University Hospital FrankfurtFrankfurt (Main)Germany
| | - Philipp Wendel
- Goethe University Frankfurt, Department of Pediatrics, Experimental Immunology and Cell TherapyFrankfurt (Main)Germany
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI)Frankfurt (Main)Germany
- Institute for Organic Chemistry and BiochemistryTechnical University of DarmstadtDarmstadtGermany
- German Cancer Consortium (DKTK)Partner Site Frankfurt/Mainz and German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Emmanuel Donnadieu
- Universite' Paris Cité, CNRS, INSERMEquipe Labellisée Ligue Contre le Cancer, Institut CochinParisFrance
| | - Martin‐Leo Hansmann
- Frankfurt Institute for Advanced Studies (FIAS)Frankfurt (Main)Germany
- Institute of General Pharmacology and ToxicologyGoethe University Frankfurt (Main)Germany
| | - Sylvia Hartmann
- Institute of PathologyUniversity Hospital Essen, University of Duisburg‐EssenEssenGermany
| | - Evelyn Ullrich
- Goethe University Frankfurt, Department of Pediatrics, Experimental Immunology and Cell TherapyFrankfurt (Main)Germany
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI)Frankfurt (Main)Germany
- Goethe University Frankfurt, University Cancer Center Frankfurt (UCT), University Hospital FrankfurtFrankfurt (Main)Germany
- German Cancer Consortium (DKTK)Partner Site Frankfurt/Mainz and German Cancer Research Center (DKFZ)HeidelbergGermany
| |
Collapse
|
3
|
Chawla SP, Pang SS, Jain D, Jeffrey S, Chawla NS, Song PY, Hall FL, Gordon EM. Gene and Cell Therapy for Sarcomas: A Review. Cancers (Basel) 2025; 17:1125. [PMID: 40227707 PMCID: PMC11987864 DOI: 10.3390/cancers17071125] [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: 01/27/2025] [Revised: 03/07/2025] [Accepted: 03/26/2025] [Indexed: 04/15/2025] Open
Abstract
Background: The heterogeneity of sarcomas and resulting distinct sub-type specific characteristics, their high recurrence rates, and tendency for distant metastasis, continue to present significant challenges to providing optimal treatments. Objective: To provide a comprehensive review of current literature and clinical trials in gene and cell therapies for sarcomas. Methods: A comprehensive literature search was conducted utilizing the following databases: PubMed, Medline, Google Scholar and clinicaltrials.gov. Search terms included "gene therapy", "cell therapy", "NK cell therapy, "CAR-T therapy", "virotherapy", "sarcoma", "gene therapy", and "solid tumors". Additional sources were identified through manual searching for references of relevant studies. No language restrictions were set. The NCT number, study status, condition, and phase were noted for clinical trials. Results: There are only three gene and cell therapies for sarcomas that have been approved by a federal regulatory agency. Rexin-G: the first tumor-targeted gene therapy vector designed to target all advanced solid malignancies, including chemo-refractory osteosarcomas and soft tissue sarcomas, was approved by the Philippine FDA in 2007. Gendicine was the first oncolytic virus approved for intratumoral delivery in China in 2003. Afami-cel, an innovative chimeric antigen receptor (CAR) T cell therapy, was approved for synovial sarcoma in the United States in 2024. Other promising therapies are discussed in the text. Conclusions: The future of gene and cell therapy for sarcomas holds great promise, as research moves to late-stage clinical development. The integration of gene and cell therapies into standard sarcoma treatment protocols has the potential to significantly improve the quality of life and outcomes for patients with this rare and challenging group of cancers.
Collapse
Affiliation(s)
- Sant P. Chawla
- Sarcoma Oncology Center/Cancer Center of Southern California, Santa Monica, CA 90403, USA; (S.P.C.); (S.S.P.); (D.J.); (S.J.); (N.S.C.)
| | - Skyler S. Pang
- Sarcoma Oncology Center/Cancer Center of Southern California, Santa Monica, CA 90403, USA; (S.P.C.); (S.S.P.); (D.J.); (S.J.); (N.S.C.)
| | - Darshit Jain
- Sarcoma Oncology Center/Cancer Center of Southern California, Santa Monica, CA 90403, USA; (S.P.C.); (S.S.P.); (D.J.); (S.J.); (N.S.C.)
| | - Samantha Jeffrey
- Sarcoma Oncology Center/Cancer Center of Southern California, Santa Monica, CA 90403, USA; (S.P.C.); (S.S.P.); (D.J.); (S.J.); (N.S.C.)
- Aveni Foundation, Santa Monica, CA 90403, USA
| | - Neal S. Chawla
- Sarcoma Oncology Center/Cancer Center of Southern California, Santa Monica, CA 90403, USA; (S.P.C.); (S.S.P.); (D.J.); (S.J.); (N.S.C.)
| | | | | | - Erlinda M. Gordon
- Sarcoma Oncology Center/Cancer Center of Southern California, Santa Monica, CA 90403, USA; (S.P.C.); (S.S.P.); (D.J.); (S.J.); (N.S.C.)
- Aveni Foundation, Santa Monica, CA 90403, USA
- Delta Next-Gene, LLC, Santa Monica, CA 90405, USA;
| |
Collapse
|
4
|
Lahouty M, Soleymanzadeh A, Kazemi S, Saadati-Maleki H, Masoudi S, Ghasemi A, Kazemi T, Mehranfar S, Fadaee M. Cell-based immunotherapy in oesophageal cancer. J Drug Target 2025:1-11. [PMID: 40063049 DOI: 10.1080/1061186x.2025.2477077] [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: 01/21/2025] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/18/2025]
Abstract
Oesophageal cancer (EC) is among the most common illnesses globally, and its prognosis is unfavourable. Surgery, radiotherapy and chemotherapy are the primary therapy options for EC. Despite the occasional efficacy of these traditional treatment modalities, individuals with EC remain at a significant risk for local recurrence and metastasis. Consequently, innovative and efficacious medicines are required. In recent decades, clinical practice has effectively implemented cell therapy, which includes both stem cell and non-stem cell-based approaches, as an innovative tumour treatment, offering renewed hope to patients with oesophageal squamous cell carcinoma (ESCC). This paper examines the theoretical framework and contemporary advancements in cell treatment for individuals with EC. We further described current clinical studies and summarised essential data related to survival and safety assessments.
Collapse
Affiliation(s)
- Masoud Lahouty
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Sama Kazemi
- Faculty of Medicine, Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Haniyeh Saadati-Maleki
- Faculty of Medicine, Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Sanaz Masoudi
- Faculty of Medicine, Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Arash Ghasemi
- Faculty of Medicine, Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Mehranfar
- Department of Genetics and Immunology, Urmia University of Medical Sciences, Urmia, Iran
| | - Manouchehr Fadaee
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
5
|
Yan X, Wang D. Innovative PBMC-derived humanized mouse model reveals CD8 + T cell-intrinsic regulation during antitumor immunity. Methods 2025; 234:286-293. [PMID: 39826657 DOI: 10.1016/j.ymeth.2025.01.011] [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: 10/30/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025] Open
Abstract
The PBMC-derived humanized mouse model (PBMC model) may serve as an excellent tool in the field of immunology for both preclinical research and personalized therapeutic strategy development. However, single transplantation of complete PBMCs without modifications prevents the identification of cell type-specific factors that are potentially involved in modulating cell-intrinsic functions for the immune response. Here, we establish an innovative strategy for PBMC model generation, where two-step transplantations coupled with cell type-specific gene manipulation were conducted to evaluate the potential role of CD8+ T cell-intrinsic factors in regulating antitumor immunity toward PDX-based tumors. This method readily yields over 10 % of human CD45+ cells within the PBMCs of humanized mice with high editing efficiency of gene expression in CD8+ T cells that can be subsequently detected in the tumor microenvironment (TME). Our work provides a new method to generate a PBMC-derived humanized mouse model for investigating regulators of interest during antitumor immunity in a cell type-specific manner.
Collapse
Affiliation(s)
- Xiaojun Yan
- State Key Laboratory of Common Mechanism Research for Major Diseases & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Donglai Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
6
|
Schoenfeld K, Habermann J, Wendel P, Harwardt J, Ullrich E, Kolmar H. T cell receptor-directed antibody-drug conjugates for the treatment of T cell-derived cancers. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200850. [PMID: 39176070 PMCID: PMC11338945 DOI: 10.1016/j.omton.2024.200850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/02/2024] [Accepted: 07/16/2024] [Indexed: 08/24/2024]
Abstract
T cell-derived cancers are hallmarked by heterogeneity, aggressiveness, and poor clinical outcomes. Available targeted therapies are severely limited due to a lack of target antigens that allow discrimination of malignant from healthy T cells. Here, we report a novel approach for the treatment of T cell diseases based on targeting the clonally rearranged T cell receptor displayed by the cancerous T cell population. As a proof of concept, we identified an antibody with unique specificity toward a distinct T cell receptor (TCR) and developed antibody-drug conjugates, precisely recognizing and eliminating target T cells while preserving overall T cell repertoire integrity and cellular immunity. Our anti-TCR antibody-drug conjugates demonstrated effective receptor-mediated cell internalization, associated with induction of cancer cell death with strong signs of apoptosis. Furthermore, cell proliferation-inhibiting bystander effects observed on target-negative cells may contribute to the molecules' anti-tumor properties precluding potential tumor escape mechanisms. To our knowledge, this represents the first anti-TCR antibody-drug conjugate designed as custom-tailored immunotherapy for T cell-driven pathologies.
Collapse
Affiliation(s)
- Katrin Schoenfeld
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Jan Habermann
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
- Goethe University, Department of Pediatrics, Experimental Immunology and Cell Therapy, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, 60596 Frankfurt am Main, Germany
| | - Philipp Wendel
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
- Goethe University, Department of Pediatrics, Experimental Immunology and Cell Therapy, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, 60596 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Julia Harwardt
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Evelyn Ullrich
- Goethe University, Department of Pediatrics, Experimental Immunology and Cell Therapy, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, 60596 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, 64283 Darmstadt, Germany
| |
Collapse
|
7
|
Long J, Wang Y, Jiang X, Ge J, Chen M, Zheng B, Wang R, Wang M, Xu M, Ke Q, Wang J. Nanomaterials Boost CAR-T Therapy for Solid Tumors. Adv Healthc Mater 2024; 13:e2304615. [PMID: 38483400 DOI: 10.1002/adhm.202304615] [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: 01/16/2024] [Revised: 02/29/2024] [Indexed: 05/22/2024]
Abstract
T cell engineering, particularly via chimeric antigen receptor (CAR) modifications for enhancing tumor specificity, has shown efficacy in treating hematologic malignancies. The extension of CAR-T cell therapy to solid tumors, however, is impeded by several challenges: The absence of tumor-specific antigens, antigen heterogeneity, a complex immunosuppressive tumor microenvironment, and physical barriers to cell infiltration. Additionally, limitations in CAR-T cell manufacturing capacity and the high costs associated with these therapies restrict their widespread application. The integration of nanomaterials into CAR-T cell production and application offers a promising avenue to mitigate these challenges. Utilizing nanomaterials in the production of CAR-T cells can decrease product variability and lower production expenses, positively impacting the targeting and persistence of CAR-T cells in treatment and minimizing adverse effects. This review comprehensively evaluates the use of various nanomaterials in the production of CAR-T cells, genetic modification, and in vivo delivery. It discusses their underlying mechanisms and potential for clinical application, with a focus on improving specificity and safety in CAR-T cell therapy.
Collapse
Affiliation(s)
- Jun Long
- Shenzhen Geim Graphene Center, Tsinghua-Berkeley Shenzhen Institute & Tsinghua Shenzhen International Graduate School, Tsinghua University, 1001 Xueyuan Road, Shenzhen, 518055, China
| | - Yian Wang
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, The Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha, 410013, China
| | - Xianjie Jiang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Junshang Ge
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, 410078, China
| | - Mingfen Chen
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Fujian Medical University, Quanzhou, 362000, China
| | - Boshu Zheng
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, No.1 Xuefu North Road University Town, Fuzhou, 350122, China
| | - Rong Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, No.1 Xuefu North Road University Town, Fuzhou, 350122, China
| | - Meifeng Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, No.1 Xuefu North Road University Town, Fuzhou, 350122, China
| | - Meifang Xu
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, No.1 Xuefu North Road University Town, Fuzhou, 350122, China
| | - Qi Ke
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, No.1 Xuefu North Road University Town, Fuzhou, 350122, China
| | - Jie Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, No.1 Xuefu North Road University Town, Fuzhou, 350122, China
| |
Collapse
|
8
|
Hosseini M, Akbari B, Shahverdi AR, Hadjati J, Faramarzi MA, Mirzaei HR, Yazdi MH. Generation of Murine Chimeric Antigen Receptor-Modified T Cells for In Vivo Studies in Syngeneic Tumor Models. Curr Protoc 2024; 4:e1107. [PMID: 39166803 DOI: 10.1002/cpz1.1107] [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: 08/23/2024]
Abstract
CAR-T cell therapy has emerged as a potent and effective tool in the immunotherapy of refractory cancers. However, challenges exist in their clinical application, necessitating extensive preclinical research to optimize their function. Various preclinical in vitro and in vivo models have been proposed for such purpose; among which immunocompetent mouse models serve as an invaluable tool in studying host immune interactions within a more realistic simulation of the tumor milieu. We hereby describe a standardized protocol for the generation of high-titer γ-retroviral vectors through transfection of the HEK293T packaging cell line. The virus-containing supernatant is further concentrated using an inhouse concentrator solution, titrated, and applied to mouse T cells purified via a convenient and rapid method by nylon-wool columns. Using the method presented here, we were able to achieve high titer γ-retrovirus and highly pure mouse T cells with desirable CAR transduction efficiency. The mouse CAR T cells produced through this protocol demonstrate favorable CAR expression and viability, thus making them suitable for further in vitro/in vivo assays. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Production of γ-retroviral vectors from retrovirus-backbone plasmids Basic Protocol 2: Concentration of γ-retrovirus-containing supernatants Basic Protocol 3: Titration of concentrated γ-retrovirus Basic Protocol 4: Isolation and activation of mouse T cells Basic Protocol 5: Transduction of activated mouse T cells, assessment of CAR expression, and expansion of CAR T cells for further in vitro/in vivo studies Support Protocol: Surface staining of cells for flow cytometric assessment of CAR expression.
Collapse
Affiliation(s)
- Mina Hosseini
- Department of Pharmaceutical Biotechnology, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnia Akbari
- Department of Medical Immunology, Tehran University of Medical Sciences, Tehran, Iran
- Vilcek Institute of Graduate Biomedical Sciences, New York University School of Medicine, New York
| | - Ahmad Reza Shahverdi
- Department of Pharmaceutical Biotechnology, Tehran University of Medical Sciences, Tehran, Iran
- Recombinant Vaccine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Jamshid Hadjati
- Department of Medical Immunology, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
9
|
Stibbs DJ, Silva Couto P, Takeuchi Y, Rafiq QA, Jackson NB, Rayat AC. Continuous manufacturing of lentiviral vectors using a stable producer cell line in a fixed-bed bioreactor. Mol Ther Methods Clin Dev 2024; 32:101209. [PMID: 38435128 PMCID: PMC10907162 DOI: 10.1016/j.omtm.2024.101209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/07/2024] [Indexed: 03/05/2024]
Abstract
Continuous manufacturing of lentiviral vectors (LVs) using stable producer cell lines could extend production periods, improve batch-to-batch reproducibility, and eliminate costly plasmid DNA and transfection reagents. A continuous process was established by expanding cells constitutively expressing third-generation LVs in the iCELLis Nano fixed-bed bioreactor. Fixed-bed bioreactors provide scalable expansion of adherent cells and enable a straightforward transition from traditional surface-based culture vessels. At 0.5 vessel volume per day (VVD), the short half-life of LVs resulted in a low total infectious titer at 1.36 × 104 TU cm-2. Higher perfusion rates increased titers, peaking at 7.87 × 104 TU cm-2 at 1.5 VVD. The supernatant at 0.5 VVD had a physical-to-infectious particle ratio of 659, whereas this was 166 ± 15 at 1, 1.5, and 2 VVD. Reducing the pH from 7.20 to 6.85 at 1.5 VVD improved the total infectious yield to 9.10 × 104 TU cm-2. Three independent runs at 1.5 VVD and a culture pH of 6.85 showed low batch-to-batch variability, with a coefficient of variation of 6.4% and 10.0% for total infectious and physical LV yield, respectively. This study demonstrated the manufacture of high-quality LV supernatant using a stable producer cell line that does not require induction.
Collapse
Affiliation(s)
- Dale J. Stibbs
- Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK
| | - Pedro Silva Couto
- Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK
| | - Yasuhiro Takeuchi
- Division of Infection and Immunity, University College London, Cruciform Building, Gower Street, London WC1E 6BT, UK
- Biotherapeutics and Advanced Therapies, Scientific Research and Innovation, Medicines and Healthcare Products Regulatory Agency, South Mimms, Potters Bar EN6 3QC, UK
| | - Qasim A. Rafiq
- Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK
| | - Nigel B. Jackson
- Cytiva, 5 Harbourgate Business Park, Southampton Road, Portsmouth PO6 4BQ, UK
| | - Andrea C.M.E. Rayat
- Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
10
|
Märkl F, Schultheiß C, Ali M, Chen SS, Zintchenko M, Egli L, Mietz J, Chijioke O, Paschold L, Spajic S, Holtermann A, Dörr J, Stock S, Zingg A, Läubli H, Piseddu I, Anz D, Minden MDV, Zhang T, Nerreter T, Hudecek M, Minguet S, Chiorazzi N, Kobold S, Binder M. Mutation-specific CAR T cells as precision therapy for IGLV3-21 R110 expressing high-risk chronic lymphocytic leukemia. Nat Commun 2024; 15:993. [PMID: 38307904 PMCID: PMC10837166 DOI: 10.1038/s41467-024-45378-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/22/2024] [Indexed: 02/04/2024] Open
Abstract
The concept of precision cell therapy targeting tumor-specific mutations is appealing but requires surface-exposed neoepitopes, which is a rarity in cancer. B cell receptors (BCR) of mature lymphoid malignancies are exceptional in that they harbor tumor-specific-stereotyped sequences in the form of point mutations that drive self-engagement of the BCR and autologous signaling. Here, we use a BCR light chain neoepitope defined by a characteristic point mutation (IGLV3-21R110) for selective targeting of a poor-risk subset of chronic lymphocytic leukemia (CLL) with chimeric antigen receptor (CAR) T cells. We develop murine and humanized CAR constructs expressed in T cells from healthy donors and CLL patients that eradicate IGLV3-21R110 expressing cell lines and primary CLL cells, but neither cells expressing the non-pathogenic IGLV3-21G110 light chain nor polyclonal healthy B cells. In vivo experiments confirm epitope-selective cytolysis in xenograft models in female mice using engrafted IGLV3-21R110 expressing cell lines or primary CLL cells. We further demonstrate in two humanized mouse models lack of cytotoxicity towards human B cells. These data provide the basis for advanced approaches of resistance-preventive and biomarker-guided cellular targeting of functionally relevant lymphoma driver mutations sparing normal B cells.
Collapse
Affiliation(s)
- Florian Märkl
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Christoph Schultheiß
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Murtaza Ali
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Shih-Shih Chen
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | | | - Lukas Egli
- Cellular Immunotherapy, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Juliane Mietz
- Cellular Immunotherapy, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Obinna Chijioke
- Cellular Immunotherapy, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Institute of Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - Lisa Paschold
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Sebastijan Spajic
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Anne Holtermann
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Janina Dörr
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Sophia Stock
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Andreas Zingg
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Cancer Immunotherapy, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Heinz Läubli
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Cancer Immunotherapy, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Ignazio Piseddu
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - David Anz
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | | | - Tianjiao Zhang
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Thomas Nerreter
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Susana Minguet
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
| | - Nicholas Chiorazzi
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Munich, Research Center for Environmental Health (HMGU), Neuherberg, Germany.
| | - Mascha Binder
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland.
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
11
|
Trainor N, Purpura KA, Middleton K, Fargo K, Hails L, Vicentini-Hogan M, McRobie C, Daniels R, Densham P, Gardin P, Fouks M, Brayer H, Malka RG, Rodin A, Ogen T, Besser MJ, Smith T, Leonard D, Bryan A. Automated production of gene-modified chimeric antigen receptor T cells using the Cocoon Platform. Cytotherapy 2023; 25:1349-1360. [PMID: 37690020 DOI: 10.1016/j.jcyt.2023.07.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/29/2023] [Accepted: 07/29/2023] [Indexed: 09/11/2023]
Abstract
Autologous cell-based therapeutics have gained increasing attention in recent years because of their efficacy at treating diseases with limited therapeutic options. Chimeric antigen receptor (CAR) T-cell therapy has demonstrated clinical success in hematologic oncology indications, providing critically ill patients with a potentially curative therapy. Although engineered cell therapies such as CAR T cells provide new options for patients with unmet needs, the high cost and complexity of manufacturing may hinder clinical and commercial translation. The Cocoon Platform (Lonza, Basel, Switzerland) addresses many challenges, such as high labor demand, process consistency, contamination risks and scalability, by enabling efficient, functionally closed and automated production, whether at clinical or commercial scale. This platform is customizable and easy to use and requires minimal operator interaction, thereby decreasing process variability. We present two processes that demonstrate the Cocoon Platform's capabilities. We employed different T-cell activation methods-OKT3 and CD3/CD28 Dynabeads (Thermo Fisher Scientific, Waltham, MA, USA)-to generate final cellular products that meet the critical quality attributes of a clinical autologous CAR T-cell product. This study demonstrates a manufacturing solution for addressing challenges with manual methods of production and facilitating the scale-up of autologous cell therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Hadar Brayer
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Rivka Gal Malka
- Wohl Institute for Translational Medicine/Advanced Biotherapy Center Good Manufacturing Practice Facility, Sheba Medical Center, Ramat Gan, Israel
| | - Anastasia Rodin
- Wohl Institute for Translational Medicine/Advanced Biotherapy Center Good Manufacturing Practice Facility, Sheba Medical Center, Ramat Gan, Israel
| | - Tal Ogen
- Wohl Institute for Translational Medicine/Advanced Biotherapy Center Good Manufacturing Practice Facility, Sheba Medical Center, Ramat Gan, Israel
| | - Michal J Besser
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat Gan, Israel; Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Davidoff Center, Rabin Medical Center, Petah Tikva, Israel
| | - Tim Smith
- Octane Biotech Inc, Kingston, Canada
| | | | - Adam Bryan
- Lonza Walkersville, Inc, Walkersville, Maryland, USA
| |
Collapse
|
12
|
Uscanga-Palomeque AC, Chávez-Escamilla AK, Alvizo-Báez CA, Saavedra-Alonso S, Terrazas-Armendáriz LD, Tamez-Guerra RS, Rodríguez-Padilla C, Alcocer-González JM. CAR-T Cell Therapy: From the Shop to Cancer Therapy. Int J Mol Sci 2023; 24:15688. [PMID: 37958672 PMCID: PMC10649325 DOI: 10.3390/ijms242115688] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/04/2023] [Accepted: 10/12/2023] [Indexed: 11/15/2023] Open
Abstract
Cancer is a worldwide health problem. Nevertheless, new technologies in the immunotherapy field have emerged. Chimeric antigen receptor (CAR) technology is a novel biological form to treat cancer; CAR-T cell genetic engineering has positively revolutionized cancer immunotherapy. In this paper, we review the latest developments in CAR-T in cancer treatment. We present the structure of the different generations and variants of CAR-T cells including TRUCK (T cells redirected for universal cytokine killing. We explain the approaches of the CAR-T cells manufactured ex vivo and in vivo. Moreover, we describe the limitations and areas of opportunity for this immunotherapy and the current challenges of treating hematological and solid cancer using CAR-T technology as well as its constraints and engineering approaches. We summarize other immune cells that have been using CAR technology, such as natural killer (NK), macrophages (M), and dendritic cells (DC). We conclude that CAR-T cells have the potential to treat not only cancer but other chronic diseases.
Collapse
Affiliation(s)
- Ashanti Concepción Uscanga-Palomeque
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66450, Nuevo León, Mexico; (A.K.C.-E.); (C.A.A.-B.); (S.S.-A.); (L.D.T.-A.); (R.S.T.-G.); (C.R.-P.)
| | | | | | | | | | | | | | - Juan Manuel Alcocer-González
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66450, Nuevo León, Mexico; (A.K.C.-E.); (C.A.A.-B.); (S.S.-A.); (L.D.T.-A.); (R.S.T.-G.); (C.R.-P.)
| |
Collapse
|
13
|
Guo C, Chen H, Yu J, Lu H, Xia Q, Li X, Guo X, Wang T, Zhi L, Niu Z, Zhu W. Engagement of an optimized lentiviral vector enhances the expression and cytotoxicity of CAR in human NK cells. Mol Immunol 2023; 155:91-99. [PMID: 36736195 DOI: 10.1016/j.molimm.2023.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 02/04/2023]
Abstract
Adoptive chimeric antigen receptor (CAR)-modified T or NK cells (CAR-T/NK) have emerged as a novel form of disease treatment. Lentiviral vectors (LVs) are commonly employed to engineer NK cells for the efficient expression of CARs. This study reported the influence of single-promoter and dual-promoter LVs on the CAR expression and cytotoxicity of engineered NK cells. We constructed a third-generation NKG2D-based CAR that kills cancer cells by targeting up to eight stress-induced ligands (NKG2DLs). Our results demonstrated that the CAR exhibits both a higher expression level and a higher coexpression concordance with the GFP reporter in HEK-293T or NK92 cells by utilizing the optimized single-promoter pCDHsp rather than the original dual-promoter pCDHdp. After puromycin selection, the pCDHsp produces robust CAR expression and enhanced in vitro cytotoxicity of engineered NK cells. Therefore, infection with a single-promoter pCDHsp lentivector is recommended to prepare CAR-engineered NK cells. This research helps to optimize the production of CAR-NK cells and enhance their functional activity, to provide CAR-NK cell products with better and more uniform quality.
Collapse
Affiliation(s)
- Changjiang Guo
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China.
| | - Han Chen
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Jie Yu
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Hui Lu
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Qing Xia
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Xiaojuan Li
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Xiali Guo
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Tong Wang
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Lingtong Zhi
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Zhiyuan Niu
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Wuling Zhu
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China.
| |
Collapse
|
14
|
Hu Y, Cao G, Chen X, Huang X, Asby N, Ankenbruck N, Rahman A, Thusu A, He Y, Riedell PA, Bishop MR, Schreiber H, Kline JP, Huang J. Antigen-Multimers: Specific, Sensitive, Precise, and Multifunctional High-Avidity CAR-Staining Reagents. MATTER 2021; 4:3917-3940. [PMID: 34901832 PMCID: PMC8654235 DOI: 10.1016/j.matt.2021.09.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Although chimeric antigen receptor (CAR) T-cell therapy has transformed cancer treatment, high-quality and universal CAR-staining reagents are urgently required to manufacture CAR T cells, predict therapy response, decipher CAR biology, and engineer new CARs. Here, we developed tetrameric and dodecameric forms of a multifunctional and extensible category of high-avidity CAR-staining reagents: antigen-multimers. Antigen-multimers detected CARs against CD19, HER2, and Tn-glycoside with significantly higher specificity, sensitivity, and precision than existing reagents. In addition to accurate CAR T-cell detection by flow cytometry, antigen-multimers also enabled ≥100-fold magnetic enrichment of rare CAR T cells, selective CAR T-cell stimulation, and high-dimensional CAR T-cell profiling by single-cell multi-omics analyses. Finally, antigen-multimers accurately captured clinical anti-CD19 CAR T cells from patients' cellular infusion products, post-infusion peripheral blood, and tumor biopsies. Antigen-multimers can be readily extended to other CAR systems by switching its antigen ligand. As such, antigen-multimers have broad clinical and research applications.
Collapse
Affiliation(s)
- Yifei Hu
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
- Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Guoshuai Cao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Xiufen Chen
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Xiaodan Huang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Nicholas Asby
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Nicholas Ankenbruck
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Ali Rahman
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Ashima Thusu
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Yanran He
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, USA
| | - Peter A. Riedell
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, USA
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL 60637, USA
| | - Michael R. Bishop
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL 60637, USA
| | - Hans Schreiber
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, USA
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL 60637, USA
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Justin P. Kline
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, USA
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL 60637, USA
| | - Jun Huang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|