1
|
Chakraborty S, Sridhar S, Ventura K, Sritharan R, Tocheva A, Sen T. Protocol to co-culture SCLC cells with human CD8 + T cells to measure tumor cell killing and T cell activation. STAR Protoc 2025; 6:103767. [PMID: 40232937 DOI: 10.1016/j.xpro.2025.103767] [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: 01/15/2025] [Revised: 02/16/2025] [Accepted: 03/20/2025] [Indexed: 04/17/2025] Open
Abstract
Small-cell lung cancer (SCLC) is an aggressive malignancy with immunosuppressive tumor microenvironment limiting immunotherapy efficacy. Here, we present a protocol to assess T cell activation and the ability of lurbinectedin to enhance anti-tumor responses using in vitro co-cultures of SCLC cells and human CD8 T cells. We describe steps for cell seeding, treatment, co-culture setup, and assessing cell viability. This protocol provides a platform to study anti-tumor T cell responses and develop new SCLC therapies. For complete details on the use and execution of the protocol, please refer to Chakraborty et al.1.
Collapse
Affiliation(s)
- Subhamoy Chakraborty
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Subhasree Sridhar
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kedwin Ventura
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ramja Sritharan
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Anna Tocheva
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Triparna Sen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
2
|
Wang Y, Jiang J, Shang K, Xu X, Sun J. Turning "trashed" genomic loci into treasurable sites for integrating chimeric antigen receptors in T and NK cells. Mol Ther 2025; 33:1368-1379. [PMID: 39980196 PMCID: PMC11997492 DOI: 10.1016/j.ymthe.2025.02.028] [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: 07/05/2024] [Revised: 10/11/2024] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
Chimeric antigen receptor (CAR)-based immune cell therapy involves genetically engineering immune cells, such as T cells and natural killer (NK) cells, to express CARs that can specifically recognize target antigens. This modification enables T/NK cells to selectively eliminate tumor cells following adoptive transfer. One common approach to stably integrate CARs into the genome of T/NK cells is through retroviral or lentiviral vectors. However, these vectors mediate semi-random gene integration, posing risks such as oncogenic mutations, gene silencing, and variable CAR expression levels. Targeted integration of CAR genes into the specific genomic locus could overcome these limitations, but identifying the optimal integration sites to maximize the safety and efficacy of CAR-T/NK cell products remains a critical question. Improper integration sites may disturb the endogenous genes surrounding the integration sites, raising safety concerns. Additionally, regulatory elements at the integration sites, such as promoters, can influence the expression level of CAR genes, thus affecting the efficacy of CAR-T/NK cells. In this review, we summarized current strategies for selecting integration sites and promoters in the engineering of CAR-T/NK cells to achieve potent anti-tumor efficacy in preclinical studies.
Collapse
Affiliation(s)
- Yajie Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China; Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China; Institute of Hematology, Zhejiang University and Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, Zhejiang, China
| | - Jie Jiang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China; Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China; Institute of Hematology, Zhejiang University and Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, Zhejiang, China
| | - Kai Shang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China; Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China; Institute of Hematology, Zhejiang University and Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, Zhejiang, China
| | - Xiaobao Xu
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China; Institute of Hematology, Zhejiang University and Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, Zhejiang, China
| | - Jie Sun
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China; Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China; Institute of Hematology, Zhejiang University and Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
3
|
Osada E, Manome Y, Sato Y. A dataset on NFKB1/NFKB2 knockout Jurkat cells and primary CD4+ T cells generated by CRISPR/Cas9 mediated gene disruption. Data Brief 2025; 59:111359. [PMID: 40027254 PMCID: PMC11870169 DOI: 10.1016/j.dib.2025.111359] [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: 12/13/2024] [Revised: 01/14/2025] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Specific biological roles of non-canonical nuclear factor-kappa B (NF-kB) signaling in human T cells remain unclear. Therefore, we genetically modified Jurkat leukemic cells using the clustered regularly interspaced palindromic repeat (CRISPR)/CRISPR-associated protein 9 (Cas9) technology to better understand the NF-kB signaling mechanisms. IKAROS family zinc finger-2 levels were downregulated in NFKB2-knocked-out cells but upregulated in NFKB1-knocked-out cells without stimulation. To further understand the biological roles of NF-kB signaling in CD4+ T cells, NFKB1/NFKB2 loci of human primary CD4+ T cells were genetically edited using the CRISPR/Cas9 technology. Quantitative polymerase chain reaction and fluorescence-activated cell sorting revealed the similar phenotypes and gene expression profiles of NFKB1/NFKB2-knocked-out human CD4+ T cells at a resting state.
Collapse
Affiliation(s)
- Erika Osada
- Core Research Facilities, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoshinobu Manome
- Core Research Facilities, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Yohei Sato
- Core Research Facilities, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
4
|
Liao Q, Xu J. Safe CAR-T: shedding light on CAR-related T-cell malignancies. EMBO Mol Med 2025; 17:589-593. [PMID: 40021932 PMCID: PMC11982530 DOI: 10.1038/s44321-025-00205-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 02/07/2025] [Accepted: 02/14/2025] [Indexed: 03/03/2025] Open
Abstract
In this Comment, J. Xu & Q. Liao propose safer genetic engineering strategies for CAR-T cells, and underscore the necessity of introducing more sensitive and reliable safety evaluation indicators, such as T cell receptor (TCR) diversity and integration site analysis.
Collapse
Affiliation(s)
- Qibin Liao
- Department of Oncology and Bio-therapeutic Center, The Third People's Hospital of Shenzhen, Second Hospital Affiliated to Southern University of Science and Technology, 518112, Shenzhen, China
| | - Jianqing Xu
- Clinical Center of Biotherapy, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.
- Institute of Biomedical Sciences, Fudan University, 200032, Shanghai, China.
- Clinical Center of Biotherapy, Zhongshan Hospital (Xiamen), Fudan University, 361015, Xiamen, China.
| |
Collapse
|
5
|
Georgiadis C, Preece R, Qasim W. Clinical development of allogeneic chimeric antigen receptor αβ-T cells. Mol Ther 2025:S1525-0016(25)00214-X. [PMID: 40156192 DOI: 10.1016/j.ymthe.2025.03.040] [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: 01/03/2025] [Revised: 03/10/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Ready-made banks of allogeneic chimeric antigen receptor (CAR) T cells, produced to be available at the time of need, offer the prospect of accessible and cost-effective cellular therapies. Various strategies have been developed to overcome allogeneic barriers, drawing on cell engineering platforms including RNA interference, protein-based restriction, and genome editing, including RNA-guided CRISPR-Cas and base editing tools. Alloreactivity and the risk of graft-versus-host disease from non-matched donor cells have been mitigated by disruption of αβ-T cell receptor expression on the surface of T cells and stringent removal of any residual αβ-T cell populations. In addition, host-mediated rejection has been tackled through a combination of augmented lymphodepletion and cell engineering strategies that have allowed infused cells to evade immune recognition or conferred resistance to lymphodepleting agents to promote persistence and expansion of effector populations. Early-phase studies using off-the-shelf universal donor CAR T cells have been undertaken mainly in the context of blood malignancies, where emerging data of clinical responses have supported wider adoption and further applications. These developments offer the prospect of alternatives to current autologous approaches through the emerging application of genome engineering solutions to improve safety, persistence, and function of universal donor products.
Collapse
Affiliation(s)
- Christos Georgiadis
- University College London Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, London WC1N 1DZ, UK
| | - Roland Preece
- University College London Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, London WC1N 1DZ, UK
| | - Waseem Qasim
- University College London Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, London WC1N 1DZ, UK.
| |
Collapse
|
6
|
Porret R, Alcaraz-Serna A, Peter B, Bernier-Latmani J, Cecchin R, Alfageme-Abello O, Ermellino L, Hafezi M, Pace E, du Pré MF, Lana E, Golshayan D, Velin D, Eyquem J, Tang Q, Petrova TV, Coukos G, Irving M, Pot C, Pantaleo G, Sollid LM, Muller YD. T cell receptor precision editing of regulatory T cells for celiac disease. Sci Transl Med 2025; 17:eadr8941. [PMID: 40106579 DOI: 10.1126/scitranslmed.adr8941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 02/11/2025] [Indexed: 03/22/2025]
Abstract
Celiac disease, a gluten-sensitive enteropathy, demonstrates a strong human leukocyte antigen (HLA) association, with more than 90% of patients carrying the HLA-DQ2.5 allotype. No therapy is available for the condition except for a lifelong gluten-free diet. To address this gap, we explored the therapeutic potential of regulatory T cells (Tregs). By orthotopic replacement of T cell receptors (TCRs) through homology-directed repair, we generated gluten-reactive HLA-DQ2.5-restricted CD4+ engineered (e) T effector cells (Teffs) and eTregs and performed in vivo experiments in HLA-DQ2.5 transgenic mice. Of five validated TCRs, TCRs specific for two immunodominant and deamidated gluten epitopes (DQ2.5-glia-α1a and DQ2.5-glia-α2) were selected for further evaluation. CD4+ eTeffs exposed to deamidated gluten through oral gavage colocalized with dendritic and B cells in the Peyer's patches and gut-draining lymph nodes and specifically migrated to the intestine. The suppressive function of human eTregs correlated with high TCR functional activity. eTregs specific for one epitope suppressed the proliferation and gut migration of CD4+ eTeffs specific for the same and the other gluten epitope, demonstrating bystander suppression. The suppression requires an antigen-specific activation of eTregs given that polyclonal Tregs failed to suppress CD4+ eTeffs. These findings highlight the potential of gluten-reactive eTregs as a therapeutic for celiac disease.
Collapse
Affiliation(s)
- Raphaël Porret
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Ana Alcaraz-Serna
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Benjamin Peter
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Jeremiah Bernier-Latmani
- Department of Oncology, Lausanne University Hospital and University of Lausanne, 1005 Lausanne, Switzerland
| | - Rebecca Cecchin
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Oscar Alfageme-Abello
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Laura Ermellino
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Morteza Hafezi
- Department of Oncology, Lausanne University Hospital and University of Lausanne, 1005 Lausanne, Switzerland
| | - Eleonora Pace
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - M Fleur du Pré
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo and Department of Immunology, Oslo University Hospital, Oslo NO-0424, Norway
| | - Erica Lana
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Dela Golshayan
- Transplantation Center, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Dominique Velin
- Service of Gastroenterology and Hepatology, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Justin Eyquem
- Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Qizhi Tang
- Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Tatiana V Petrova
- Department of Oncology, Lausanne University Hospital and University of Lausanne, 1005 Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, Lausanne University Hospital and University of Lausanne, 1005 Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne Branch, 1066 Lausanne, Switzerland
| | - Melita Irving
- Department of Oncology, Lausanne University Hospital and University of Lausanne, 1005 Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne Branch, 1066 Lausanne, Switzerland
| | - Caroline Pot
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Giuseppe Pantaleo
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Ludvig M Sollid
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo and Department of Immunology, Oslo University Hospital, Oslo NO-0424, Norway
| | - Yannick D Muller
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
- Centre for Human Immunology Lausanne, Lausanne CH-1005, Switzerland
| |
Collapse
|
7
|
Lennerz V, Doppler C, Fatho M, Dröge A, Schaper S, Gennermann K, Genzel N, Plassmann S, Weismann D, Lukowski SW, Bents D, Beushausen C, Kriese K, Herbst H, Seitz V, Hammer R, Adam PJ, Eggeling S, Wölfel C, Wölfel T, Hennig S. T-cell receptors identified by a personalized antigen-agnostic screening approach target shared neoantigen KRAS Q61H. Front Immunol 2025; 16:1509855. [PMID: 40165973 PMCID: PMC11955635 DOI: 10.3389/fimmu.2025.1509855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Adoptive cell therapy (ACT) with TCR-engineered T-cells represents a promising alternative to TIL- or CAR-T therapies for patients with advanced solid cancers. Currently, selection of therapeutic TCRs critically depends on knowing the target antigens, a condition excluding most patients from treatment. Direct antigen-agnostic identification of tumor-specific T-cell clonotypes and TCR-T manufacturing using their TCRs can advance ACT for patients with aggressive solid cancers. We present a method to identify tumor-specific clonotypes from surgical specimens by comparing TCRβ-chain repertoires of TILs and adjacent tissue-resident lymphocytes. In six out of seven NSCLC-patients analyzed, our selection of tumor-specific clonotypes based on TIL-abundance and high tumor-to-nontumor frequency ratios was confirmed by gene expression signatures determined by scRNA-Seq. In three patients, we demonstrated that predicted tumor-specific clonotypes reacted against autologous tumors. For one of these patients, we engineered TCR-T cells with four candidate tumor-specific TCRs that showed reactivity against the patient's tumor and HLA-matched NSCLC cell lines. The TCR-T cells were then used to screen for candidate neoantigens and aberrantly expressed antigens. Three TCRs recognized recurrent driver-mutation KRAS Q61H-peptide ILDTAGHEEY presented by HLA-A*01:01. The TCRs were also dominant in a tumor relapse, one was found in cell free DNA. The finding of homologous TCRs in independent KRAS Q61H-positive cancers suggests a therapeutic opportunity for HLA-matched patients with KRAS Q61H-expressing tumors.
Collapse
MESH Headings
- Humans
- Proto-Oncogene Proteins p21(ras)/genetics
- Proto-Oncogene Proteins p21(ras)/immunology
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/therapy
- Lung Neoplasms/genetics
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Immunotherapy, Adoptive/methods
- Precision Medicine/methods
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Female
- Male
- Cell Line, Tumor
- Aged
- Mutation
Collapse
Affiliation(s)
- Volker Lennerz
- Internal Medicine III, University Medical Center (UMC) of the Johannes Gutenberg University Mainz, Mainz, Germany
- HSDiagnomics GmbH, Berlin, Germany
- TheryCell GmbH, Berlin, Germany
| | - Christoph Doppler
- Internal Medicine III, University Medical Center (UMC) of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Martina Fatho
- Internal Medicine III, University Medical Center (UMC) of the Johannes Gutenberg University Mainz, Mainz, Germany
| | | | | | | | | | | | - David Weismann
- Boehringer Ingelheim RCV, GmbH & Co KG., Cancer Immunology & Immune Modulation, Vienna, Austria
| | - Samuel W. Lukowski
- Boehringer Ingelheim RCV, GmbH & Co KG., Cancer Immunology & Immune Modulation, Vienna, Austria
| | | | | | - Karen Kriese
- Vivantes Pathology, Vivantes Clinic Neukölln, Berlin, Germany
| | - Hermann Herbst
- Vivantes Pathology, Vivantes Clinic Neukölln, Berlin, Germany
| | | | - Rudolf Hammer
- HSDiagnomics GmbH, Berlin, Germany
- TheryCell GmbH, Berlin, Germany
| | - Paul J. Adam
- Boehringer Ingelheim RCV, GmbH & Co KG., Cancer Immunology & Immune Modulation, Vienna, Austria
| | - Stephan Eggeling
- Vivantes Clinic Neukölln, Vivantes Thoracic Surgery, Berlin, Germany
| | - Catherine Wölfel
- Internal Medicine III, University Medical Center (UMC) of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Thomas Wölfel
- Internal Medicine III, University Medical Center (UMC) of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Steffen Hennig
- HSDiagnomics GmbH, Berlin, Germany
- TheryCell GmbH, Berlin, Germany
| |
Collapse
|
8
|
Tommasi A, Cappabianca D, Bugel M, Gimse K, Lund-Peterson K, Shrestha H, Arutyunov D, Williams JA, Police SR, Indurthi V, Davis SZ, Murtaza M, Capitini CM, Saha K. Efficient nonviral integration of large transgenes into human T cells using Cas9-CLIPT. Mol Ther Methods Clin Dev 2025; 33:101437. [PMID: 40123742 PMCID: PMC11930092 DOI: 10.1016/j.omtm.2025.101437] [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: 06/17/2024] [Accepted: 02/17/2025] [Indexed: 03/25/2025]
Abstract
CRISPR-Cas9 ribonucleoproteins (RNPs) combined with a nucleic acid template encoding a chimeric antigen receptor (CAR) transgene can edit human cells to produce CAR T cells with precise CAR insertion at a single locus. However, many human cells have adverse innate immune responses to foreign nucleic acids, particularly circular double-stranded DNA (dsDNA). Here, we introduce Cleaved, LInearized with Protein Template (Cas9-CLIPT), a circular plasmid containing a single target sequence for the Cas9 RNP, such that during manufacturing, Cas9-RNP binds and cleaves the plasmid to linearize the dsDNA in vitro. Cas9-RNP remains bound to the linearized template and is delivered to cells to promote precise knock-in via homology-directed repair with Cas9-CLIPT. Cas9-CLIPT Nanoplasmids generate up to 1.7-fold higher rates of precise knock-in relative to linearized dsDNA, reaching efficiencies up to 60% with non-homologous end joining inhibition. Cas9-CLIPT-manufactured GD2 TRAC-CAR T cells are potent against GD2+ neuroblastoma cells and exhibit an enriched stem cell memory phenotype. On several electroporation instruments and approaching clinically relevant yields, we successfully manufactured TRAC-CAR T cells using Cas9-CLIPT plasmids containing large (2-6 kb) transgenes. Cas9-CLIPT strategies have the potential to simplify donor template production and integrate large transgenes, allowing for more efficient nonviral manufacturing of multifunctional, genome-edited immune cell therapies.
Collapse
Affiliation(s)
- Anna Tommasi
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Dan Cappabianca
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Madison Bugel
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Kirstan Gimse
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | | | | | | | | | | | | | - Sage Z. Davis
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
| | - Muhammed Murtaza
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- Department of Surgery, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Christian M. Capitini
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Krishanu Saha
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
9
|
Luo C, Zhang R, Guo R, Wu L, Xue T, He Y, Jin Y, Zhao Y, Zhang Z, Zhang P, Ye S, Li X, Li D, Zhang W, Wang C, Lai L, Pan-Hammarström Q, Wucherpfennig KW, Gao Z, Pan D, Zeng Z. Integrated computational analysis identifies therapeutic targets with dual action in cancer cells and T cells. Immunity 2025; 58:745-765.e9. [PMID: 40023158 DOI: 10.1016/j.immuni.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 10/11/2024] [Accepted: 02/04/2025] [Indexed: 03/04/2025]
Abstract
Many cancer drugs that target cancer cell pathways also impair the immune system. We developed a computational target discovery platform to enable examination of both cancer and immune cells so as to identify pathways that restrain tumor progression and potentiate anti-tumor immunity. Immune-related CRISPR screen analyzer of functional targets (ICRAFT) integrates immune-related CRISPR screen datasets, single-cell RNA sequencing (scRNA-seq) data, and pre-treatment RNA-seq data from clinical trials, enabling a systems-level approach to therapeutic target discovery. Using ICRAFT, we identified numerous targets that enhance both cancer cell susceptibility to immune attack and T cell activation, including tumor necrosis factor (TNF) alpha-induced protein 3 (TNFAIP3), protein tyrosine phosphatase non-receptor type 2 (PTPN2), and suppressor of cytokine signaling 1 (SOCS1). In cancer cells, Tnfaip3 (A20) deletion activated the TNF-nuclear factor kappa-B (NF-κB) pathway, promoting chemokine expression and T cell recruitment to the tumor. T cell-mediated elimination of Tnaifp3-null cancer cells was primarily driven by TNF-induced apoptosis. Inactivation of Tnfaip3 in T cells enhanced anti-tumor efficacy. By integrating diverse functional genomics and clinical datasets, ICRAFT provides an interactive resource toward a deeper understanding of anti-tumor immunity and immuno-oncology drug development.
Collapse
Affiliation(s)
- Ce Luo
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China
| | - Rui Zhang
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China
| | - Rui Guo
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China
| | - Lijian Wu
- School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Teng Xue
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan 610213, China
| | - Yufeng He
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China
| | - Yiteng Jin
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China
| | - Yanping Zhao
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zongxu Zhang
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China
| | - Peng Zhang
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China
| | - Sitong Ye
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA; Yale School of Medicine, New Haven, CT 06510, USA
| | - Xiaohong Li
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China
| | - Dian Li
- Division of Biology and Biomedical Sciences, Washington University in St. Louis School of Medicine, Saint Louis, MO 63108, USA
| | - Wubing Zhang
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Chenfei Wang
- Shanghai Putuo District People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Luhua Lai
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China; Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan 610213, China
| | - Qiang Pan-Hammarström
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm 17165, Sweden
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02215, USA
| | - Zhidong Gao
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing 100084, China.
| | - Deng Pan
- School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China.
| | - Zexian Zeng
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China; Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan 610213, China.
| |
Collapse
|
10
|
Du W, Noyan F, McCallion O, Drosdek V, Kath J, Glaser V, Fuster-Garcia C, Yang M, Stein M, Franke C, Pu Y, Weber O, Polansky JK, Cathomen T, Jaeckel E, Hester J, Issa F, Volk HD, Schmueck-Henneresse M, Reinke P, Wagner DL. Gene editing of CD3 epsilon to redirect regulatory T cells for adoptive T cell transfer. Mol Ther 2025; 33:997-1013. [PMID: 39905729 PMCID: PMC11897813 DOI: 10.1016/j.ymthe.2025.01.045] [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: 06/02/2024] [Revised: 12/20/2024] [Accepted: 01/30/2025] [Indexed: 02/06/2025] Open
Abstract
Adoptive transfer of antigen-specific regulatory T cells (Tregs) is a promising strategy to combat immunopathologies in transplantation and autoimmune diseases. However, their low frequency in peripheral blood poses challenges for both manufacturing and clinical application. Chimeric antigen receptors have been used to redirect the specificity of Tregs, using retroviral vectors. However, retroviral gene transfer is costly, time consuming, and raises safety issues. Here, we explored non-viral CRISPR-Cas12a gene editing to redirect Tregs, using human leukocyte antigen (HLA)-A2-specific constructs for proof-of-concept studies in transplantation models. Knock-in of an antigen-binding domain into the N terminus of CD3 epsilon (CD3ε) gene generates Tregs expressing a chimeric CD3ε-T cell receptor fusion construct (TRuC) protein that integrates into the endogenous TCR/CD3 complex. These CD3ε-TRuC Tregs exhibit potent antigen-dependent activation while maintaining responsiveness to TCR/CD3 stimulation. This enables preferential enrichment of TRuC-redirected Tregs over CD3ε knockout Tregs via repetitive CD3/CD28 stimulation in a good manufacturing practice-compatible expansion system. CD3ε-TRuC Tregs retained their phenotypic, epigenetic, and functional identity. In a humanized mouse model, HLA-A2-specific CD3ε-TRuC Tregs demonstrate superior protection of allogeneic HLA-A2+ skin grafts from rejection compared with polyclonal Tregs. This approach provides a pathway for developing clinical-grade CD3ε-TRuC-based Treg cell products for transplantation immunotherapy and other immunopathologies.
Collapse
Affiliation(s)
- Weijie Du
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), 13353 Berlin, Germany; BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Fatih Noyan
- Department of Gastroenterology, Infectious Diseases and Hepatology & Endocrinology, Hannover Medical School, Hannover, Germany
| | - Oliver McCallion
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Science, University of Oxford, Oxford OX3 9DU, UK
| | - Vanessa Drosdek
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), 13353 Berlin, Germany; BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Jonas Kath
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), 13353 Berlin, Germany; BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Viktor Glaser
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), 13353 Berlin, Germany; BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Carla Fuster-Garcia
- Institute for Transfusion Medicine and Gene Therapy, Medical Center, University of Freiburg, 79106 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Mingxing Yang
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Maik Stein
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), 13353 Berlin, Germany; BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Clemens Franke
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), 13353 Berlin, Germany; BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Yaolin Pu
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), 13353 Berlin, Germany; BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Olaf Weber
- Institute of Molecular Medicine and Experimental Immunology (IMMEI), University of Bonn, Biomedical Center II, Venusberg Campus 1, 53127 Bonn, Germany
| | - Julia K Polansky
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; German Rheumatism Research Centre, Deutsches Rheuma-Forschungszentrum, ein Leibniz Institut, Berlin, Germany
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center, University of Freiburg, 79106 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Elmar Jaeckel
- Department of Gastroenterology, Infectious Diseases and Hepatology & Endocrinology, Hannover Medical School, Hannover, Germany; Department of Liver Transplantation, Multi Organ Transplant Program, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Joanna Hester
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Science, University of Oxford, Oxford OX3 9DU, UK
| | - Fadi Issa
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Science, University of Oxford, Oxford OX3 9DU, UK
| | - Hans-Dieter Volk
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), 13353 Berlin, Germany; BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Michael Schmueck-Henneresse
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), 13353 Berlin, Germany; BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Petra Reinke
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), 13353 Berlin, Germany; Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Dimitrios L Wagner
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), 13353 Berlin, Germany; BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Institute of Transfusion Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany; Center for Cell and Gene Therapy, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
11
|
Shankar K, Zingler-Hoslet I, Tabima DM, Zima S, Shi L, Gimse K, Forsberg MH, Katta V, Davis SZ, Maldonado D, Russell BE, Murtaza M, Tsai SQ, Ayuso JM, Capitini CM, Saha K. Virus-free CRISPR knockin of a chimeric antigen receptor into KLRC1 generates potent GD2-specific natural killer cells. Mol Ther 2025; 33:1014-1030. [PMID: 39815622 PMCID: PMC11897758 DOI: 10.1016/j.ymthe.2025.01.024] [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: 02/05/2024] [Revised: 08/09/2024] [Accepted: 01/10/2025] [Indexed: 01/18/2025] Open
Abstract
Natural killer (NK) cells are an appealing off-the-shelf, allogeneic cellular therapy due to their cytotoxic profile. However, their activity against solid tumors remains suboptimal in part due to the upregulation of NK-inhibitory ligands, such as HLA-E, within the tumor microenvironment. Here, we utilize CRISPR-Cas9 to disrupt the KLRC1 gene (encoding the HLA-E-binding NKG2A receptor) and perform non-viral insertion of a GD2-targeting chimeric antigen receptor (CAR) within NK cells isolated from human peripheral blood. Genome editing with CRISPR-Cas9 ribonucleoprotein complexes yields efficient genomic disruption of the KLRC1 gene with 98% knockout efficiency and specific knockin of the GD2 CAR transgene as high as 23%, with minimal off-target activity as shown by CHANGE-seq, in-out PCR, amplicon sequencing, and long-read whole-genome sequencing. KLRC1-GD2 CAR NK cells display high viability and proliferation, as well as precise cellular targeting and potency against GD2+ human tumor cells. Notably, KLRC1-GD2 CAR NK cells overcome HLA-E-based inhibition in vitro against HLA-E-expressing, GD2+ melanoma cells. Using a single-step, virus-free genome editing workflow, this study demonstrates the feasibility of precisely disrupting inhibitory signaling within NK cells via CRISPR-Cas9 while expressing a CAR to generate potent allogeneic cell therapies against HLA-E+ solid tumors.
Collapse
Affiliation(s)
- Keerthana Shankar
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Isabelle Zingler-Hoslet
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Diana M Tabima
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Seth Zima
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Dermatology, University of Wisconsin-Madison, Madison, WI 53715, USA; Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Lei Shi
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53715, USA; Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Kirstan Gimse
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Matthew H Forsberg
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53715, USA; Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Varun Katta
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sage Z Davis
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Daniel Maldonado
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Brittany E Russell
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Muhammed Murtaza
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Surgery, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Shengdar Q Tsai
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jose M Ayuso
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Dermatology, University of Wisconsin-Madison, Madison, WI 53715, USA; Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Christian M Capitini
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53715, USA
| | - Krishanu Saha
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53715, USA.
| |
Collapse
|
12
|
Walsh ZH, Shah P, Kothapalli N, Shah SB, Nikolenyi G, Brodtman DZ, Leuzzi G, Rogava M, Mu M, Ho P, Abuzaid S, Vasan N, AlQuraishi M, Milner JD, Ciccia A, Melms JC, Izar B. Mapping variant effects on anti-tumor hallmarks of primary human T cells with base-editing screens. Nat Biotechnol 2025; 43:384-395. [PMID: 38783148 DOI: 10.1038/s41587-024-02235-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/10/2024] [Indexed: 05/25/2024]
Abstract
Single-nucleotide variants (SNVs) in key T cell genes can drive clinical pathologies and could be repurposed to improve cellular cancer immunotherapies. Here, we perform massively parallel base-editing screens to generate thousands of variants at gene loci annotated with known or potential clinical relevance. We discover a broad landscape of putative gain-of-function (GOF) and loss-of-function (LOF) mutations, including in PIK3CD and the gene encoding its regulatory subunit, PIK3R1, LCK, SOS1, AKT1 and RHOA. Base editing of PIK3CD and PIK3R1 variants in T cells with an engineered T cell receptor specific to a melanoma epitope or in different generations of CD19 chimeric antigen receptor (CAR) T cells demonstrates that discovered GOF variants, but not LOF or silent mutation controls, enhanced signaling, cytokine production and lysis of cognate melanoma and leukemia cell models, respectively. Additionally, we show that generations of CD19 CAR T cells engineered with PIK3CD GOF mutations demonstrate enhanced antigen-specific signaling, cytokine production and leukemia cell killing, including when benchmarked against other recent strategies.
Collapse
Affiliation(s)
- Zachary H Walsh
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Parin Shah
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Neeharika Kothapalli
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Shivem B Shah
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Gergo Nikolenyi
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - D Zack Brodtman
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Giuseppe Leuzzi
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, USA
| | - Meri Rogava
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Michael Mu
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Patricia Ho
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Sinan Abuzaid
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Neil Vasan
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Mohammed AlQuraishi
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Joshua D Milner
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Alberto Ciccia
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, USA
| | - Johannes C Melms
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Benjamin Izar
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA.
- Columbia Center for Translational Immunology, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
13
|
Li K, Wu Y, Hu Y, Yue Y, Ma X, Wang S, Gan X, Xu X. Bipolar Electrode-Based Precise Manipulation and Selective Electroporation of Cells. Anal Chem 2025; 97:3908-3919. [PMID: 39950441 DOI: 10.1021/acs.analchem.4c05041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
The intracellular delivery toward a specific type of single cell shows great potential in single-cell-specific therapeutic and diagnostic applications. Most of the current methods require high-precision micromanipulators or require multiple steps for motor fabrication, which hinders their practical application. Herein, we for the first time report a method for precise manipulation and selective electroporation of cells using a bipolar electrode. We achieved the precise control of the position of target cells via dielectrophoresis (DEP) at the edge of a bipolar electrode and selective electroporation of specific cells by the local intensified electric field obtained by the gap between the driving electrodes under a direct current (DC) pulse train. Active cell targeting and electroporation of cells are demonstrated using a rotating electric field to drive the cells and a train of pulses to transfect the cells. By harnessing pDEP and twDEP, our device offers the ability to precisely control the movement and placement of specific cells under a rotating electric field and enables the targeted cells to be driven toward regions where the electric field strength is optimized for efficient electroporation. Our method was demonstrated to be applicable across a wide range of cell types, by selective electroporation of different cells including yeast cells, K562 cells, THP-1 cells, 293T cells, and SNU-1 cells. In addition to the injection of fluorescence dye molecules, we also further demonstrated the introduction of plasmids into the SNU-1 cells successfully. This approach is generic and applicable to bacteria and a wide range of cell types, offering an important and novel experimental tool for targeted delivery and single-cell analysis.
Collapse
Affiliation(s)
- Kemu Li
- School of Integrated Circuits, Southeast University, Sipailou 2, Nanjing 210096, P. R. China
- School of Microelectronics, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, P. R. China
| | - Yupan Wu
- School of Integrated Circuits, Southeast University, Sipailou 2, Nanjing 210096, P. R. China
- School of Microelectronics, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, P. R. China
- Yulin Environmental Monitoring Station, Yulin, Shanxi 719000, P. R. China
- Yangtze River Delta Research Institute of NPU, Taicang, Suzhou, Jiangsu 215400, P. R. China
| | - Yan Hu
- The First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Taicang, Suzhou, Jiangsu 215400, P. R. China
| | - Yuanbo Yue
- School of Microelectronics, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, P. R. China
| | - Xun Ma
- School of Microelectronics, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, P. R. China
| | - Shaoxi Wang
- School of Microelectronics, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, P. R. China
| | - Xuetao Gan
- School of Microelectronics, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, P. R. China
| | - Xiaohui Xu
- The First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Taicang, Suzhou, Jiangsu 215400, P. R. China
| |
Collapse
|
14
|
Gowen BG, Melton K, Leong WI, Khekare P, McCawley S, Chan J, Boivin P, Jani V, Cantor AJ, Tambe A, Haak-Frendscho M, Janatpour MJ, Wei SC. Systematic identification and characterization of high efficiency Cas9 guide RNAs for therapeutic targeting of ADAR. PLoS One 2025; 20:e0317745. [PMID: 39992915 PMCID: PMC11849909 DOI: 10.1371/journal.pone.0317745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 01/03/2025] [Indexed: 02/26/2025] Open
Abstract
Therapeutic targeting of the adenosine deaminase ADAR has great potential in cancer and other indications; however, it remains unclear what approach can enable effective and selective therapeutic inhibition. Herein, we conduct multi-staged guide RNA screening and identify high efficiency Cas9 guide RNAs to enable a CRISPR/Cas-based approach for ADAR knockout. Through characterization in human primary immune cell systems we observe similar activity with two-part guide RNA and single guide RNA, dose responsive activity, similar guide activity rank order across different cell types, and favorable computational off-target profiles of candidate guide RNAs. We determine that knockout of ADAR using these guide RNAs induces pharmacodynamic responses primarily consisting of immunological responses such as a type I interferon response, consistent with the known function of ADAR as a key regulator of dsRNA sensing. We observe similar biological effects with targeting only the p150 isoform or both p110 and p150 isoforms of ADAR, indicating that at least in the contexts evaluated, loss of p150 ADAR mediates the primary response. These findings provide a resource of well-characterized, high efficiency ADAR-targeting Cas9 guide RNAs suitable for genomic medicines utilizing different delivery modalities and addressing different therapeutic areas.
Collapse
Affiliation(s)
- Benjamin G. Gowen
- Spotlight Therapeutics, Hayward, California, United States of America
| | - Kory Melton
- Spotlight Therapeutics, Hayward, California, United States of America
| | - Weng In Leong
- Spotlight Therapeutics, Hayward, California, United States of America
| | - Prachi Khekare
- Spotlight Therapeutics, Hayward, California, United States of America
| | - Shannon McCawley
- Spotlight Therapeutics, Hayward, California, United States of America
| | - Jean Chan
- Spotlight Therapeutics, Hayward, California, United States of America
| | - Pierre Boivin
- Spotlight Therapeutics, Hayward, California, United States of America
| | - Vihasi Jani
- Spotlight Therapeutics, Hayward, California, United States of America
| | - Aaron J. Cantor
- Spotlight Therapeutics, Hayward, California, United States of America
| | - Akshay Tambe
- Spotlight Therapeutics, Hayward, California, United States of America
| | | | - Mary J. Janatpour
- Spotlight Therapeutics, Hayward, California, United States of America
| | - Spencer C. Wei
- Spotlight Therapeutics, Hayward, California, United States of America
| |
Collapse
|
15
|
Perico L, Casiraghi F, Benigni A, Remuzzi G. Is there a place for engineered immune cell therapies in autoimmune diseases? Trends Mol Med 2025:S1471-4914(25)00011-5. [PMID: 39984382 DOI: 10.1016/j.molmed.2025.01.011] [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: 11/19/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 02/23/2025]
Abstract
The ability to engineer immune cells yielded a transformative era in oncology. Early clinical trials demonstrated the efficacy of chimeric antigen receptor (CAR) T cells in resetting the immune system, motivating the expansion of this treatment beyond cancer, including autoimmune conditions. In this review, we discuss the current state of CAR T cell research in autoimmune diseases, examining the main challenges that limit widespread adoption of this therapy, such as complex isolation protocols, stringent immunosuppression, risk of secondary malignancies, and variable efficacy. We also review the studies addressing these limitations by development of off-the-shelf allogeneic CAR T cells, tunable safety systems, and antigen-specific therapies, which hold the potential to improve safety and accessibility of this treatment in clinical practice.
Collapse
Affiliation(s)
- Luca Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy.
| | | | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| |
Collapse
|
16
|
Džafo E, Hafezi M, Attianese GMPG, Reichenbach P, Grillet S, Garcia H, Cribioli E, Voize C, Tissot S, de Silly RV, Coukos G, Scholten K, Irving M, Gentner B. DNA-dependent protein kinase inhibitors PI-103 and samotolisib augment CRISPR/Cas9 knock-in efficiency in human T cells. Cytotherapy 2025:S1465-3249(25)00059-3. [PMID: 40100189 DOI: 10.1016/j.jcyt.2025.02.001] [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: 10/14/2024] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 03/20/2025]
Abstract
The adoptive transfer of autologous peripheral blood T cells gene-modified to express preselected, tumor antigen-specific T-cell receptors (TCRs) is a promising treatment for solid cancers. While gene-transfer by viral transduction is highly efficient, the insertional site is not targeted and persistence of the T cells is oftentimes limited. In contrast, site-specific integration of the TCR into the TCR α chain (TRAC) locus by CRISPR/Cas9 has been shown to enable more consistent and physiologic levels of exogenous TCR expression coupled with superior persistence and tumor control in preclinical studies. Here, we sought to improve the efficiency of CRISPR/Cas9 mediated TCR knock-in (KI) into the TRAC locus of primary human T cells. In addition to the previously reported DNA-dependent protein kinase (DNA-PK) inhibitor M3814, we demonstrated that PI-103 and samotolisib markedly increase KI efficiency in a process that is good manufacturing process (GMP)-compatible. Importantly, samotolisib enabled the generation of a potent T-cell product, having no negative impact on T-cell viability, phenotype, expansion, effector function, and tumor control. Overall, we conclude that our GMP-compatible CRISPR/Cas9 protocol comprising samotolisib to augment TCR KI efficiency is suitable for the generation of genetically modified T cells for clinical use.
Collapse
Affiliation(s)
- Emina Džafo
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL), and Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Morteza Hafezi
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL), and Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Greta Maria Paola Giordano Attianese
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL), and Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Patrick Reichenbach
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL), and Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Stephane Grillet
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL), and Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Hélène Garcia
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL), and Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Elisabetta Cribioli
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL), and Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Christel Voize
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL), and Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Stephanie Tissot
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL), and Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Romain Vuillefroy de Silly
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL), and Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - George Coukos
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL), and Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Kirsten Scholten
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL), and Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Melita Irving
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL), and Lausanne University Hospital (CHUV), Lausanne, Switzerland.
| | - Bernhard Gentner
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, University of Lausanne (UNIL), and Lausanne University Hospital (CHUV), Lausanne, Switzerland
| |
Collapse
|
17
|
Chang CR, Vykunta VS, Lee JHJ, Li K, Kochendoerfer C, Muldoon JJ, Wang CH, Mazumder T, Sun Y, Goodman DB, Nyberg WA, Liu C, Allain V, Rothrock A, Ye CJ, Marson A, Shy BR, Eyquem J. SEED-Selection enables high-efficiency enrichment of primary T cells edited at multiple loci. Nat Biotechnol 2025:10.1038/s41587-024-02531-6. [PMID: 39910194 DOI: 10.1038/s41587-024-02531-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 12/10/2024] [Indexed: 02/07/2025]
Abstract
Engineering T cell specificity and function at multiple loci can generate more effective cellular therapies, but current manufacturing methods produce heterogenous mixtures of partially engineered cells. Here we develop a one-step process to enrich unlabeled cells containing knock-ins at multiple target loci using a family of repair templates named synthetic exon expression disruptors (SEEDs). SEEDs associate transgene integration with the disruption of a paired target endogenous surface protein while preserving target expression in nonmodified and partially edited cells to enable their removal (SEED-Selection). We design SEEDs to modify three critical loci encoding T cell specificity, coreceptor expression and major histocompatibility complex expression. The results demonstrate up to 98% purity after selection for individual modifications and up to 90% purity for six simultaneous edits (three knock-ins and three knockouts). This method is compatible with existing clinical manufacturing workflows and can be readily adapted to other loci to facilitate production of complex gene-edited cell therapies.
Collapse
Affiliation(s)
- Christopher R Chang
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Vivasvan S Vykunta
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Jae Hyun J Lee
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Ke Li
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Clara Kochendoerfer
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Joseph J Muldoon
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Charlotte H Wang
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Thomas Mazumder
- Division of Rheumatology, University of California, San Francisco, San Francisco, CA, USA
| | - Yang Sun
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Division of Rheumatology, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel B Goodman
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - William A Nyberg
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Chang Liu
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Vincent Allain
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Université Paris Cité, INSERM UMR976, Hôpital Saint-Louis, Paris, France
| | - Allison Rothrock
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Chun J Ye
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Division of Rheumatology, University of California, San Francisco, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
- Arc Institute, Palo Alto, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Alexander Marson
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Brian R Shy
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA.
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA.
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| | - Justin Eyquem
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA.
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA.
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA.
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA.
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
18
|
Zhang D, Duque-Jimenez J, Facchinetti F, Brixi G, Rhee K, Feng WW, Jänne PA, Zhou X. Transferrin receptor targeting chimeras for membrane protein degradation. Nature 2025; 638:787-795. [PMID: 39322661 PMCID: PMC11839386 DOI: 10.1038/s41586-024-07947-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/14/2024] [Indexed: 09/27/2024]
Abstract
Cancer cells require high levels of iron for rapid proliferation, leading to significant upregulation of cell-surface transferrin receptor 1 (TfR1), which mediates iron uptake by binding to the iron-carrying protein transferrin1-3. Leveraging this phenomenon and the fast endocytosis rate of TfR1 (refs. 4,5), we developed transferrin receptor targeting chimeras (TransTACs), a heterobispecific antibody modality for membrane protein degradation. TransTACs are engineered to drive rapid co-internalization of a target protein of interest and TfR1 from the cell surface, and to enable target protein entry into the lysosomal degradation pathway. We show that TransTACs can efficiently degrade a diverse range of single-pass, multi-pass, native or synthetic membrane proteins, including epidermal growth factor receptor, programmed cell death 1 ligand 1, cluster of differentiation 20 and chimeric antigen receptor. In example applications, TransTACs enabled the reversible control of human primary chimeric antigen receptor T cells and the targeting of drug-resistant epidermal growth factor receptor-driven lung cancer with the exon 19 deletion/T790M/C797S mutations in a mouse xenograft model. TransTACs represent a promising new family of bifunctional antibodies for precise manipulation of membrane proteins and targeted cancer therapy.
Collapse
Affiliation(s)
- Dingpeng Zhang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | | | - Francesco Facchinetti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Kaitlin Rhee
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - William W Feng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Pasi A Jänne
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Xin Zhou
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
19
|
Liao S, Li X, Lu Y, Luo K. Nanomedicine in Immunotherapy for Non-Small Cell Lung Cancer: Applications and Perspectives. SMALL METHODS 2025:e2401783. [PMID: 39871783 DOI: 10.1002/smtd.202401783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/17/2025] [Indexed: 01/29/2025]
Abstract
Non-small cell lung cancer (NSCLC) has a strikingly high incidence rate globally. Although immunotherapy brings a great breakthrough in its clinical treatment of NSCLC, significant challenges still need to be overcome. The development of novel multi-functional nanomedicines in the realm of tumor immunotherapy offers promising opportunities for NSCLC patients, as nanomedicines exhibit significant advantages, including specific targeting of tumor cells, improved drug bioavailability, reduced systemic toxicity, and overcoming of immune resistance. In this review, the core features and current clinical status of strategies for NSCLC immunotherapy including immune checkpoint blockade, antibody-drug conjugates, cell engagers, adoptive cells, and cancer vaccines, are surveyed. Particular emphasis is placed on the recent development of nanomedicines that boost these strategies. Nanomedicine can provide novel perspectives for NSCLC immunotherapy.
Collapse
Affiliation(s)
- Shuangsi Liao
- Division of Thoracic Tumor Multimodality Treatment, Department of Radiation Oncology, Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Cancer Center, Breast Center, Institute of Breast Health Medicine, Laboratory of Clinical Cell Therapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaoling Li
- Division of Thoracic Tumor Multimodality Treatment, Department of Radiation Oncology, Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Cancer Center, Breast Center, Institute of Breast Health Medicine, Laboratory of Clinical Cell Therapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - You Lu
- Division of Thoracic Tumor Multimodality Treatment, Department of Radiation Oncology, Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Cancer Center, Breast Center, Institute of Breast Health Medicine, Laboratory of Clinical Cell Therapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kui Luo
- Division of Thoracic Tumor Multimodality Treatment, Department of Radiation Oncology, Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Cancer Center, Breast Center, Institute of Breast Health Medicine, Laboratory of Clinical Cell Therapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
20
|
Coren LV, Trivett MT, Welker JL, Thomas JA, Gorelick RJ, Kose E, Immonen TT, Czarra K, Fennessey CM, Trubey CM, Lifson JD, Swanstrom AE. Modifications to rhesus macaque TCR constant regions improve TCR cell surface expression. PLoS One 2025; 20:e0314751. [PMID: 39787126 PMCID: PMC11717225 DOI: 10.1371/journal.pone.0314751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/17/2024] [Indexed: 01/12/2025] Open
Abstract
T cell immunotherapy success is dependent on effective levels of antigen receptor expressed at the surface of engineered cells. Efforts to optimize surface expression in T cell receptor (TCR)-based therapeutic approaches include optimization of cellular engineering methods and coding sequences, and reducing the likelihood of exogenous TCR α and β chains mispairing with the endogenous TCR chains. Approaches to promote correct human TCR chain pairing include constant region mutations to create an additional disulfide bond between the two chains, full murinization of the constant region of the TCR α and β sequences, and a minimal set of murine mutations to the TCR α and β constant regions. Preclinical animal models are valuable tools to optimize engineering designs and methods, and to evaluate the potential for off-target tissue injury. To further develop rhesus macaque models for TCR based cellular immunotherapy, we tested methods for improving cell surface expression of rhesus macaque TCR in rhesus macaque primary cells by generating five alternative TCRαβ constant region constructs in the context of a SIV Gag-specific TCR: 1. human codon optimized rhesus macaque (RH); 2. RH TCR with an additional disulfide linkage; 3. rhesus macaque constant sequences with minimal murine amino acid substitutions; 4. murinized constant sequences; and 5. murinized constant sequences with a portion of the exposed FG loop in the β constant sequence replaced with rhesus macaque sequence to reduce potential immunogencity. Murinization or mutation of a minimal set of amino acids to the corresponding murine sequence of the constant region resulted in the greatest increase in rhesus macaque TCR surface expression relative to wild type. All novel TCR constructs retained the ability to induce production of cytokines in response to cognate peptide antigen specific stimulation. This work can inform the design of TCRs selected for use in rhesus macaque models of TCR-based cellular immunotherapy.
Collapse
MESH Headings
- Animals
- Macaca mulatta
- Humans
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Mice
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Lori V. Coren
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Matthew T. Trivett
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Jorden L. Welker
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - James A. Thomas
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Robert J. Gorelick
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Emek Kose
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Taina T. Immonen
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Kelli Czarra
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Christine M. Fennessey
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Charles M. Trubey
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Adrienne E. Swanstrom
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| |
Collapse
|
21
|
Callegari A, Kueblbeck M, Morero NR, Serrano-Solano B, Ellenberg J. Rapid generation of homozygous fluorescent knock-in human cells using CRISPR-Cas9 genome editing and validation by automated imaging and digital PCR screening. Nat Protoc 2025; 20:26-66. [PMID: 39304762 DOI: 10.1038/s41596-024-01043-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 06/26/2024] [Indexed: 09/22/2024]
Abstract
We previously described a protocol for genome engineering of mammalian cultured cells with clustered regularly interspaced short palindromic repeats and associated protein 9 (CRISPR-Cas9) to generate homozygous knock-ins of fluorescent tags into endogenous genes. Here we are updating this former protocol to reflect major improvements in the workflow regarding efficiency and throughput. In brief, we have improved our method by combining high-efficiency electroporation of optimized CRISPR-Cas9 reagents, screening of single cell-derived clones by automated bright-field and fluorescence imaging, rapidly assessing the number of tagged alleles and potential off-targets using digital polymerase chain reaction (PCR) and automated data analysis. Compared with the original protocol, our current procedure (1) substantially increases the efficiency of tag integration, (2) automates the identification of clones derived from single cells with correct subcellular localization of the tagged protein and (3) provides a quantitative and high throughput assay to measure the number of on- and off-target integrations with digital PCR. The increased efficiency of the new procedure reduces the number of clones that need to be analyzed in-depth by more than tenfold and yields to more than 26% of homozygous clones in polyploid cancer cell lines in a single genome engineering round. Overall, we were able to dramatically reduce the hands-on time from 30 d to 10 d during the overall ~10 week procedure, allowing a single person to process up to five genes in parallel, assuming that validated reagents-for example, PCR primers, digital PCR assays and western blot antibodies-are available.
Collapse
Affiliation(s)
- Andrea Callegari
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Institute of Biophysics, Ulm University, Ulm, Germany
| | - Moritz Kueblbeck
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Anavo Therapeutics GmbH, Heidelberg, Germany
| | - Natalia Rosalía Morero
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Beatriz Serrano-Solano
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Euro-Bioimaging ERIC Bio-Hub, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Jan Ellenberg
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
22
|
Li Z, Ma Y, Li C, Xiao S, Liang H. Photo-Cross-linked DNA Structures Greatly Improves Their Serum Nuclease Resistances and Gene Knock-In Efficiencies. SMALL METHODS 2024:e2401346. [PMID: 39713911 DOI: 10.1002/smtd.202401346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/10/2024] [Indexed: 12/24/2024]
Abstract
The stabilization and structural integrity of DNA architectures remain significant challenges in their biomedical applications, particularly when inserting functional units into the genome using long single-stranded DNA (lssDNA). To address these challenges, a site-specific photo-cross-linking method is employed. Single-stranded oligonucleotides, containing one or two photosensitive cyanovinylcarbazole nucleoside (CNVK) molecules, are precisely incorporated and cross-linked at the specific sites of ssDNA through base-pairing, followed by rapid UV irradiation at 365 nm. This interstrand photo-cross-linking improves the thermal stability of DNA duplexes and allows this study to afford a tetrahedral DNA nanostructure in a yield of >94%. Most importantly, the photo-cross-linked DNA architectures exhibit high resistances against serum degradation, especially prevent digestion of exonuclease III (exo III), which is common in conventional lambda-processing method. Meanwhile, this photo-cross-linking treatment can significantly improve the knock-in (KI) efficiencies of lssDNA in different cells including 293T, K562, and HepG2, approximately three to eightfold those of the uncross-linked lssDNA, and remain a low cytotoxicity. Given the significantly enhanced nuclease resistance in serum and improved KI efficiencies, this study anticipates that this photo-cross-linking method will become a valuable tool in technologically advanced biomedical applications, such as nanotechnology and nucleic acid therapy.
Collapse
Affiliation(s)
- Zhigang Li
- Hefei National Research Center for Physical Sciences at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Youwei Ma
- Institute of Materials, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, 1015, Switzerland
| | - Chengxu Li
- Key Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Shiyan Xiao
- Hefei National Research Center for Physical Sciences at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Haojun Liang
- Hefei National Research Center for Physical Sciences at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
23
|
Gehrke L, Gonçalves VDR, Andrae D, Rasko T, Ho P, Einsele H, Hudecek M, Friedel SR. Current Non-Viral-Based Strategies to Manufacture CAR-T Cells. Int J Mol Sci 2024; 25:13685. [PMID: 39769449 PMCID: PMC11728233 DOI: 10.3390/ijms252413685] [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: 11/19/2024] [Revised: 12/12/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
The successful application of CAR-T cells in the treatment of hematologic malignancies has fundamentally changed cancer therapy. With increasing numbers of registered CAR-T cell clinical trials, efforts are being made to streamline and reduce the costs of CAR-T cell manufacturing while improving their safety. To date, all approved CAR-T cell products have relied on viral-based gene delivery and genomic integration methods. While viral vectors offer high transfection efficiencies, concerns regarding potential malignant transformation coupled with costly and time-consuming vector manufacturing are constant drivers in the search for cheaper, easier-to-use, safer, and more efficient alternatives. In this review, we examine different non-viral gene transfer methods as alternatives for CAR-T cell production, their advantages and disadvantages, and examples of their applications. Transposon-based gene transfer methods lead to stable but non-targeted gene integration, are easy to handle, and achieve high gene transfer rates. Programmable endonucleases allow targeted integration, reducing the potential risk of integration-mediated malignant transformation of CAR-T cells. Non-integrating CAR-encoding vectors avoid this risk completely and achieve only transient CAR expression. With these promising alternative techniques for gene transfer, all avenues are open to fully exploiting the potential of next-generation CAR-T cell therapy and applying it in a wide range of applications.
Collapse
Affiliation(s)
- Leon Gehrke
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Vasco Dos Reis Gonçalves
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Dominik Andrae
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Tamas Rasko
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Patrick Ho
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Hermann Einsele
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
- Fraunhofer-Institut für Zelltherapie und Immunologie, Außenstelle Zelluläre Immuntherapie, 97070 Würzburg, Germany
| | - Sabrina R. Friedel
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
24
|
Di Carlo E, Sorrentino C. State of the art CRISPR-based strategies for cancer diagnostics and treatment. Biomark Res 2024; 12:156. [PMID: 39696697 DOI: 10.1186/s40364-024-00701-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) technology is a groundbreaking and dynamic molecular tool for DNA and RNA "surgery". CRISPR/Cas9 is the most widely applied system in oncology research. It is a major advancement in genome manipulation due to its precision, efficiency, scalability and versatility compared to previous gene editing methods. It has shown great potential not only in the targeting of oncogenes or genes coding for immune checkpoint molecules, and in engineering T cells, but also in targeting epigenomic disturbances, which contribute to cancer development and progression. It has proven useful for detecting genetic mutations, enabling the large-scale screening of genes involved in tumor onset, progression and drug resistance, and in speeding up the development of highly targeted therapies tailored to the genetic and immunological profiles of the patient's tumor. Furthermore, the recently discovered Cas12 and Cas13 systems have expanded Cas9-based editing applications, providing new opportunities in the diagnosis and treatment of cancer. In addition to traditional cis-cleavage, they exhibit trans-cleavage activity, which enables their use as sensitive and specific diagnostic tools. Diagnostic platforms like DETECTR, which employs the Cas12 enzyme, that cuts single-stranded DNA reporters, and SHERLOCK, which uses Cas12, or Cas13, that specifically target and cleave single-stranded RNA, can be exploited to speed up and advance oncological diagnostics. Overall, CRISPR platform has the great potential to improve molecular diagnostics and the functionality and safety of engineered cellular medicines. Here, we will emphasize the potentially transformative impact of CRISPR technology in the field of oncology compared to traditional treatments, diagnostic and prognostic approaches, and highlight the opportunities and challenges raised by using the newly introduced CRISPR-based systems for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Emma Di Carlo
- Department of Medicine and Sciences of Aging, "G. d'Annunzio University" of Chieti- Pescara, Via dei Vestini, Chieti, 66100, Italy.
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Via L. Polacchi 11, Chieti, 66100, Italy.
| | - Carlo Sorrentino
- Department of Medicine and Sciences of Aging, "G. d'Annunzio University" of Chieti- Pescara, Via dei Vestini, Chieti, 66100, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Via L. Polacchi 11, Chieti, 66100, Italy
| |
Collapse
|
25
|
Ursch LT, Müschen JS, Ritter J, Klermund J, Bernard BE, Kolb S, Warmuth L, Andrieux G, Miller G, Jiménez-Muñoz M, Theis FJ, Boerries M, Busch DH, Cathomen T, Schumann K. Modulation of TCR stimulation and pifithrin-α improve the genomic safety profile of CRISPR-engineered human T cells. Cell Rep Med 2024; 5:101846. [PMID: 39637860 PMCID: PMC11722128 DOI: 10.1016/j.xcrm.2024.101846] [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: 02/05/2024] [Revised: 06/24/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024]
Abstract
CRISPR-engineered chimeric antigen receptor (CAR) T cells are at the forefront of novel cancer treatments. However, several reports describe the occurrence of CRISPR-induced chromosomal aberrations. So far, measures to increase the genomic safety of T cell products focused mainly on the components of the CRISPR-Cas9 system and less on T cell-intrinsic features, such as their massive expansion after T cell receptor (TCR) stimulation. Here, we describe driving forces of indel formation in primary human T cells. Increased T cell activation and proliferation speed correlate with larger deletions. Editing of non-activated T cells reduces the risk of large deletions with the downside of reduced knockout efficiencies. Alternatively, the addition of the small-molecule pifithrin-α limits large deletions, chromosomal translocations, and aneuploidy in a p53-independent manner while maintaining the functionality of CRISPR-engineered T cells, including CAR T cells. Controlling T cell activation and pifithrin-α treatment are easily implementable strategies to improve the genomic integrity of CRISPR-engineered T cells.
Collapse
Affiliation(s)
- Laurenz T Ursch
- Technical University of Munich (TUM), School of Medicine and Health, Department of Preclinical Medicine, Institute for Medical Microbiology, Immunology and Hygiene, 81675 Munich, Germany
| | - Jule S Müschen
- Technical University of Munich (TUM), School of Medicine and Health, Department of Preclinical Medicine, Institute for Medical Microbiology, Immunology and Hygiene, 81675 Munich, Germany
| | - Julia Ritter
- Technical University of Munich (TUM), School of Medicine and Health, Department of Preclinical Medicine, Institute for Medical Microbiology, Immunology and Hygiene, 81675 Munich, Germany
| | - Julia Klermund
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, 79106 Freiburg, Germany; Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, 79106 Freiburg, Germany
| | - Bettina E Bernard
- Technical University of Munich (TUM), School of Medicine and Health, Department of Preclinical Medicine, Institute for Medical Microbiology, Immunology and Hygiene, 81675 Munich, Germany
| | - Saskia Kolb
- Technical University of Munich (TUM), School of Medicine and Health, Department of Preclinical Medicine, Institute for Medical Microbiology, Immunology and Hygiene, 81675 Munich, Germany
| | - Linda Warmuth
- Technical University of Munich (TUM), School of Medicine and Health, Department of Preclinical Medicine, Institute for Medical Microbiology, Immunology and Hygiene, 81675 Munich, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Freiburg, Germany; Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Gregor Miller
- Core Facility Statistical Consulting, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Marina Jiménez-Muñoz
- Core Facility Statistical Consulting, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; School of Computing, Information and Technology, Technical University of Munich, 85748 Garching, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Freiburg, Germany; Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, a partnership between DKFZ and Medical Center - University of Freiburg, 79106 Freiburg, Germany
| | - Dirk H Busch
- Technical University of Munich (TUM), School of Medicine and Health, Department of Preclinical Medicine, Institute for Medical Microbiology, Immunology and Hygiene, 81675 Munich, Germany; German Center for Infection Research, Deutsches Zentrum für Infektionsforschung (DZIF), Partner Site Munich, 81675 Munich, Germany
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, 79106 Freiburg, Germany; Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Kathrin Schumann
- Technical University of Munich (TUM), School of Medicine and Health, Department of Preclinical Medicine, Institute for Medical Microbiology, Immunology and Hygiene, 81675 Munich, Germany; TUM, Institute for Advanced Study, 85748 Garching, Germany.
| |
Collapse
|
26
|
Westhaus A, Barba-Sarasua E, Chen Y, Hsu K, Scott S, Knight M, Haase F, Mesa Mora S, Houghton BC, Roca-Pinilla R, Kalajdzic P, O'Neill G, Thrasher AJ, Santilli G, Lisowski L. Tailoring capsid-directed evolution technology for improved AAV-mediated CAR-T generation. Mol Ther 2024:S1525-0016(24)00811-6. [PMID: 39673125 DOI: 10.1016/j.ymthe.2024.12.012] [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: 03/25/2024] [Revised: 10/31/2024] [Accepted: 12/10/2024] [Indexed: 12/16/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell (CAR-T) therapies present options for patients diagnosed with certain leukemias. Recent advances of the technology included a method to integrate the CAR into the T cell receptor alpha constant (TRAC) locus to take advantage of the endogenous promoter and regulatory elements for CAR expression. This method used adeno-associated viral (AAV) vectors based on AAV6 to deliver the donor template encoding the CAR construct. Since the original publication, improvements have been made to this targeted CAR integration technique; however, none of those techniques focused on improving the AAV vector used to deliver the therapeutic cargo. The herein presented study developed a novel AAV capsid directed evolution platform that allows for specifically selecting for novel AAV capsid variants that enable more efficient targeted gene editing-mediated CAR construct integration into the TRAC locus in primary T cells. Using this new platform, we selected several novel AAVs that enable more efficient editing in T cells than AAV6. Two novel capsids, AAV-T1 and AAV-T2, were able to mediate 5-fold improvement for on-target knockin, which resulted in 5-fold reduction of the vector dose to produce highly cytolytic T cells against a brain tumor cell line.
Collapse
Affiliation(s)
- Adrian Westhaus
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia; Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College, London, UK
| | - Elena Barba-Sarasua
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Yuyan Chen
- Children's Cancer Research Unit, Kids Research, The Children's Hospital at Westmead, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Kenneth Hsu
- Children's Cancer Research Unit, Kids Research, The Children's Hospital at Westmead, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Suzanne Scott
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia; Australian e-Health Research Centre, Commonwealth Scientific and Industrial Research Organisation, Sydney, NSW, Australia
| | - Maddison Knight
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Florencia Haase
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Santiago Mesa Mora
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Benjamin C Houghton
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College, London, UK
| | - Ramon Roca-Pinilla
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Predrag Kalajdzic
- Vector and Genome Engineering Facility, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Geraldine O'Neill
- Children's Cancer Research Unit, Kids Research, The Children's Hospital at Westmead, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Adrian J Thrasher
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College, London, UK
| | - Giorgia Santilli
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College, London, UK
| | - Leszek Lisowski
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia; Australian Genome Therapeutics Centre, Children's Medical Research Institute and Sydney Children's Hospitals Network, Westmead, NSW 2145, Australia; Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine - National Research Institute, Warsaw, Poland.
| |
Collapse
|
27
|
Xie K, Starzyk J, Majumdar I, Wang J, Rincones K, Tran T, Lee D, Niemi S, Famiglietti J, Suter B, Shan R, Wu H. Efficient non-viral immune cell engineering using circular single-stranded DNA-mediated genomic integration. Nat Biotechnol 2024:10.1038/s41587-024-02504-9. [PMID: 39663372 DOI: 10.1038/s41587-024-02504-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 11/12/2024] [Indexed: 12/13/2024]
Abstract
The use of adeno-associated viruses (AAVs) as donors for homology-directed repair (HDR)-mediated genome engineering is limited by safety issues, manufacturing constraints and restricted packaging limits. Non-viral targeted genetic knock-ins rely primarily on double-stranded DNA (dsDNA) and linear single-stranded DNA (lssDNA) donors. dsDNA is known to have low efficiency and high cytotoxicity, while lssDNA is challenging for scaled manufacture. In this study, we developed a non-viral genome writing catalyst (GATALYST) system that allows production of circular single-stranded DNAs (cssDNAs) up to approximately 20 kilobases as donor templates for highly efficient precision transgene integration. cssDNA donors enable knock-in efficiency of up to 70% in induced pluripotent stem cells (iPSCs) and improved efficiency in multiple clinically relevant primary immune cell types and at multiple genomic loci implicated for clinical applications with various nuclease editor systems. The high precision and efficiency in chimeric antigen receptor (CAR)-T and natural killer (NK) cells, improved safety, payload flexibility and scalable manufacturability of cssDNA shows potential for future applications of genome engineering.
Collapse
Affiliation(s)
- Keqiang Xie
- Full Circles Therapeutics, Cambridge, MA, USA.
| | | | | | - Jiao Wang
- Full Circles Therapeutics, Cambridge, MA, USA
| | | | - Thao Tran
- Full Circles Therapeutics, Cambridge, MA, USA
| | - Danna Lee
- Full Circles Therapeutics, Cambridge, MA, USA
| | - Sarah Niemi
- Full Circles Therapeutics, Cambridge, MA, USA
| | | | | | - Richard Shan
- Full Circles Therapeutics, Cambridge, MA, USA
- Stellate DNA, Cambridge, MA, USA
| | - Hao Wu
- Full Circles Therapeutics, Cambridge, MA, USA.
- Stellate DNA, Cambridge, MA, USA.
| |
Collapse
|
28
|
Wang J, Caimi PF. CAR assembly line: Taking CAR T-cell manufacturing to the next level. Best Pract Res Clin Haematol 2024; 37:101595. [PMID: 40074509 DOI: 10.1016/j.beha.2024.101595] [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: 05/31/2024] [Accepted: 12/04/2024] [Indexed: 03/14/2025]
Abstract
The widespread adoption of chimeric antigen receptor (CAR) T-cell therapy has been limited by complex, resource-intensive manufacturing processes. This review discusses the latest innovations aiming to improve and streamline CAR T-cell production across key steps like T-cell activation, genetic modification, expansion, and scaling. Promising techniques highlighted include generating CAR T cells from non-activated lymphocytes to retain a stem-like phenotype and function, non-viral gene transfer leveraging platforms like transposon and CRISPR, all-in-one fully automated bioreactors like the CliniMACS Prodigy and the Lonza Cocoon, rapid CAR T-cell manufacturing via abbreviating or eliminating ex vivo T-cell culture, implementing decentralized point-of-care automated manufacturing platforms, and optimizing centralized bioreactor infrastructure integrating end-to-end automation. Adoption of these emerging technologies can reduce production costs and timelines while enhancing product quality and accessibility. However, significant knowledge gaps persist regarding the feasibility, superiority, and optimal protocols for effectively incorporating many emerging techniques into widespread clinical practice. Further validation through clinical studies is still needed for many of these novel approaches.
Collapse
Affiliation(s)
- Jiasheng Wang
- Department of Hematology/Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH, USA.
| | - Paolo F Caimi
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
29
|
Webber BR, Johnson MJ, Skeate JG, Slipek NJ, Lahr WS, DeFeo AP, Mills LJ, Qiu X, Rathmann B, Diers MD, Wick B, Henley T, Choudhry M, Starr TK, McIvor RS, Moriarity BS. Cas9-induced targeted integration of large DNA payloads in primary human T cells via homology-mediated end-joining DNA repair. Nat Biomed Eng 2024; 8:1553-1570. [PMID: 38092857 PMCID: PMC11169092 DOI: 10.1038/s41551-023-01157-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 11/02/2023] [Indexed: 01/12/2024]
Abstract
The reliance on viral vectors for the production of genetically engineered immune cells for adoptive cellular therapies remains a translational bottleneck. Here we report a method leveraging the DNA repair pathway homology-mediated end joining, as well as optimized reagent composition and delivery, for the Cas9-induced targeted integration of large DNA payloads into primary human T cells with low toxicity and at efficiencies nearing those of viral vectors (targeted knock-in of 1-6.7 kb payloads at rates of up to 70% at multiple targeted genomic loci and with cell viabilities of over 80%). We used the method to produce T cells with an engineered T-cell receptor or a chimaeric antigen receptor and show that the cells maintained low levels of exhaustion markers and excellent capacities for proliferation and cytokine production and that they elicited potent antitumour cytotoxicity in vitro and in mice. The method is readily adaptable to current good manufacturing practices and scale-up processes, and hence may be used as an alternative to viral vectors for the production of genetically engineered T cells for cancer immunotherapies.
Collapse
Affiliation(s)
- Beau R Webber
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA.
| | - Matthew J Johnson
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Joseph G Skeate
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Nicholas J Slipek
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Walker S Lahr
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Anthony P DeFeo
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Lauren J Mills
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Xiaohong Qiu
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Blaine Rathmann
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Miechaleen D Diers
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Bryce Wick
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
| | | | | | - Timothy K Starr
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
- Department of Ob-Gyn and Women's Health, University of Minnesota, Minneapolis, MN, USA
| | - R Scott McIvor
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - Branden S Moriarity
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
30
|
Lei T, Wang Y, Zhang Y, Yang Y, Cao J, Huang J, Chen J, Chen H, Zhang J, Wang L, Xu X, Gale RP, Wang L. Leveraging CRISPR gene editing technology to optimize the efficacy, safety and accessibility of CAR T-cell therapy. Leukemia 2024; 38:2517-2543. [PMID: 39455854 PMCID: PMC11588664 DOI: 10.1038/s41375-024-02444-y] [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: 04/19/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024]
Abstract
Chimeric Antigen Receptor (CAR)-T-cell therapy has revolutionized cancer immune therapy. However, challenges remain including increasing efficacy, reducing adverse events and increasing accessibility. Use of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) technology can effectively perform various functions such as precise integration, multi-gene editing, and genome-wide functional regulation. Additionally, CRISPR screening using large-scale guide RNA (gRNA) genetic perturbation provides an unbiased approach to understanding mechanisms underlying anti-cancer efficacy of CAR T-cells. Several emerging CRISPR tools with high specificity, controllability and efficiency are useful to modify CAR T-cells and identify new targets. In this review we summarize potential uses of the CRISPR system to improve results of CAR T-cells therapy including optimizing efficacy and safety and, developing universal CAR T-cells. We discuss challenges facing CRISPR gene editing and propose solutions highlighting future research directions in CAR T-cell therapy.
Collapse
Affiliation(s)
- Tao Lei
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Yazhuo Wang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuchen Zhang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Yufei Yang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Jiaying Cao
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Jiansong Huang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Jiali Chen
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Huajing Chen
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Jiayi Zhang
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Luzheng Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Robert Peter Gale
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College of Science, Technology and Medicine, London, UK.
| | - Liang Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
31
|
James SE, Chen S, Ng BD, Fischman JS, Jahn L, Boardman AP, Rajagopalan A, Elias HK, Massa A, Manuele D, Nichols KB, Lazrak A, Lee N, Roche AM, McFarland AG, Petrichenko A, Everett JK, Bushman FD, Fei T, Kousa AI, Lemarquis AL, DeWolf S, Peled JU, Vardhana SA, Klebanoff CA, van den Brink MRM. Leucine zipper-based immunomagnetic purification of CAR T cells displaying multiple receptors. Nat Biomed Eng 2024; 8:1592-1614. [PMID: 39715901 PMCID: PMC11917073 DOI: 10.1038/s41551-024-01287-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/26/2024] [Indexed: 12/25/2024]
Abstract
Resistance to chimaeric antigen receptor (CAR) T cell therapy develops through multiple mechanisms, most notably antigen loss and tumour-induced immune suppression. It has been suggested that T cells expressing multiple CARs may overcome the resistance of tumours and that T cells expressing receptors that switch inhibitory immune-checkpoint signals into costimulatory signals may enhance the activity of the T cells in the tumour microenvironment. However, engineering multiple features into a single T cell product is difficult because of the transgene-packaging constraints of current gene-delivery vectors. Here we describe a cell-sorting method that leverages leucine zippers for the selective single-step immunomagnetic purification of cells co-transduced with two vectors. Such 'Zip sorting' facilitated the generation of T cells simultaneously expressing up to four CARs and coexpressing up to three 'switch' receptors. In syngeneic mouse models, T cells with multiple CARs and multiple switch receptors eliminated antigenically heterogeneous populations of leukaemia cells coexpressing multiple inhibitory ligands. By combining diverse therapeutic strategies, Zip-sorted multi-CAR multi-switch-receptor T cells can overcome multiple mechanisms of CAR T cell resistance.
Collapse
Affiliation(s)
- Scott E James
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA.
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- City of Hope National Medical Center, Duarte, CA, USA.
| | - Sophia Chen
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- City of Hope National Medical Center, Duarte, CA, USA
| | - Brandon D Ng
- Weill Cornell Medical College, New York, NY, USA
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
| | - Jacob S Fischman
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Lorenz Jahn
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
| | - Alexander P Boardman
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Adhithi Rajagopalan
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- City of Hope National Medical Center, Duarte, CA, USA
| | - Harold K Elias
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alyssa Massa
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- City of Hope National Medical Center, Duarte, CA, USA
| | - Dylan Manuele
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
| | | | - Amina Lazrak
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
| | - Nicole Lee
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
| | - Aoife M Roche
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander G McFarland
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Angelina Petrichenko
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John K Everett
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Teng Fei
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anastasia I Kousa
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- City of Hope National Medical Center, Duarte, CA, USA
| | - Andri L Lemarquis
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- City of Hope National Medical Center, Duarte, CA, USA
| | - Susan DeWolf
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan U Peled
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Santosha A Vardhana
- Weill Cornell Medical College, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christopher A Klebanoff
- Weill Cornell Medical College, New York, NY, USA
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marcel R M van den Brink
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA.
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
32
|
Kapetanovic E, Weber CR, Bruand M, Pöschl D, Kucharczyk J, Hirth E, Dietsche C, Khan R, Wagner B, Belli O, Vazquez-Lombardi R, Castellanos-Rueda R, Di Roberto RB, Kalinka K, Raess L, Ly K, Rai S, Dittrich PS, Platt RJ, Oricchio E, Reddy ST. Engineered allogeneic T cells decoupling T-cell-receptor and CD3 signalling enhance the antitumour activity of bispecific antibodies. Nat Biomed Eng 2024; 8:1665-1681. [PMID: 39322719 PMCID: PMC11668682 DOI: 10.1038/s41551-024-01255-x] [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: 03/08/2023] [Accepted: 08/09/2024] [Indexed: 09/27/2024]
Abstract
Bispecific antibodies (biAbs) used in cancer immunotherapies rely on functional autologous T cells, which are often damaged and depleted in patients with haematological malignancies and in other immunocompromised patients. The adoptive transfer of allogeneic T cells from healthy donors can enhance the efficacy of biAbs, but donor T cells binding to host-cell antigens cause an unwanted alloreactive response. Here we show that allogeneic T cells engineered with a T-cell receptor that does not convert antigen binding into cluster of differentiation 3 (CD3) signalling decouples antigen-mediated T-cell activation from T-cell cytotoxicity while preserving the surface expression of the T-cell-receptor-CD3 signalling complex as well as biAb-mediated CD3 signalling and T-cell activation. In mice with CD19+ tumour xenografts, treatment with the engineered human cells in combination with blinatumomab (a clinically approved biAb) led to the recognition and clearance of tumour cells in the absence of detectable alloreactivity. Our findings support the development of immunotherapies combining biAbs and 'off-the-shelf' allogeneic T cells.
Collapse
MESH Headings
- Antibodies, Bispecific/pharmacology
- Antibodies, Bispecific/immunology
- Animals
- Humans
- CD3 Complex/immunology
- CD3 Complex/metabolism
- T-Lymphocytes/immunology
- Signal Transduction/drug effects
- Mice
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Lymphocyte Activation/immunology
- Lymphocyte Activation/drug effects
- Cell Line, Tumor
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Cell Engineering/methods
- Antigens, CD19/immunology
- Antigens, CD19/metabolism
- Xenograft Model Antitumor Assays
- Allogeneic Cells/immunology
Collapse
Affiliation(s)
- Edo Kapetanovic
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Cédric R Weber
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Marine Bruand
- Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
- School of Life Sciences, EPFL, Lausanne, Switzerland
- Swiss Cancer Center Leman, Lausanne, Switzerland
| | - Daniel Pöschl
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Jakub Kucharczyk
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Elisabeth Hirth
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Claudius Dietsche
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Riyaz Khan
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Bastian Wagner
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Olivier Belli
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | | | - Rocío Castellanos-Rueda
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Life Science Zurich Graduate School, Systems Biology, ETH Zurich, University of Zurich, Zurich, Switzerland
| | - Raphael B Di Roberto
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Kevin Kalinka
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Luca Raess
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Kevin Ly
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Shivam Rai
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Petra S Dittrich
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Randall J Platt
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Elisa Oricchio
- Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
- School of Life Sciences, EPFL, Lausanne, Switzerland
- Swiss Cancer Center Leman, Lausanne, Switzerland
| | - Sai T Reddy
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland.
| |
Collapse
|
33
|
Yoo SP, Yuan X, Engstrom C, Chang P, Li S, Lathrop L, Lagosh J, Seet C, Kohn DB, Crooks GM. Stage-specific CAR-mediated signaling generates naïve-like, TCR-null CAR T cells from induced pluripotent stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.25.624041. [PMID: 39651198 PMCID: PMC11623545 DOI: 10.1101/2024.11.25.624041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Genetically modified, induced pluripotent stem cells (iPSCs) offer a promising allogeneic source for the generation of functionally enhanced, chimeric antigen receptor (CAR) T cells. However, the signaling of CARs during early T cell development and the removal of the endogenous T cell receptor required to prevent alloreactivity pose significant challenges to the production of mature conventional CAR T cells from iPSCs. Here, we show that TCR-null, CD8αβ CAR T cells can be efficiently generated from iPSCs by engineering stage-specific onset of CAR expression and signaling to both permit conventional T cell development and to induce efficient positive selection. CAR T cells produced using this approach displayed a uniform, naïve T cell phenotype and demonstrated superior antigen-specific cytotoxicity compared to iPSC-derived effector memory CAR T cells. Multimodal sequencing revealed CAR-mediated positive selection induced the persistent upregulation of key transcription factors involved in naïve T cell development. Achieving precise control of CAR expression and signaling in developmentally sensitive T precursors will be critical to realizing the full potential for "off-the-shelf", iPSC-derived cellular therapies.
Collapse
|
34
|
Amiri M, Moaveni AK, Majidi Zolbin M, Shademan B, Nourazarian A. Optimizing cancer treatment: the synergistic potential of CAR-T cell therapy and CRISPR/Cas9. Front Immunol 2024; 15:1462697. [PMID: 39582866 PMCID: PMC11581867 DOI: 10.3389/fimmu.2024.1462697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/23/2024] [Indexed: 11/26/2024] Open
Abstract
Optimizing cancer treatment has become a pivotal goal in modern oncology, with advancements in immunotherapy and genetic engineering offering promising avenues. CAR-T cell therapy, a revolutionary approach that harnesses the body's own immune cells to target and destroy cancer cells, has shown remarkable success, particularly in treating acute lymphoblastic leukemia (ALL), and in treating other hematologic malignancies. While CAR-T cell therapy has shown promise, challenges such as high cost and manufacturing complexity remain. However, its efficacy in solid tumors remains limited. The integration of CRISPR/Cas9 technology, a powerful and precise genome-editing tool, also raises safety concerns regarding unintended edits and off-target effects, offers a synergistic potential to overcome these limitations. CRISPR/Cas9 can enhance CAR-T cell therapy by improving the specificity and persistence of CAR-T cells, reducing off-target effects, and engineering resistance to tumor-induced immunosuppression. This combination can also facilitate the knockout of immune checkpoint inhibitors, boosting the anti-tumor activity of CAR-T cells. Recent studies have demonstrated that CRISPR/Cas9-edited CAR-T cells can target previously untreatable cancer types, offering new hope for patients with refractory cancers. This synergistic approach not only enhances the efficacy of cancer treatment but also paves the way for personalized therapies tailored to individual genetic profiles. This review highlights the ongoing research efforts to refine this approach and explores its potential to revolutionize cancer treatment across a broader range of malignancies. As research progresses, the integration of CAR-T cell therapy and CRISPR/Cas9 holds the promise of transforming cancer treatment, making it more effective and accessible. This review explores the current advancements, challenges, and future prospects of this innovative therapeutic strategy.
Collapse
Affiliation(s)
- Maryam Amiri
- Pediatric Urology and Regenerative Medicine Research Center, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Kian Moaveni
- Pediatric Urology and Regenerative Medicine Research Center, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Majidi Zolbin
- Pediatric Urology and Regenerative Medicine Research Center, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Behrouz Shademan
- Medical Journalism, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| |
Collapse
|
35
|
Kleinboehl EW, Laoharawee K, Jensen JD, Peterson JJ, Slipek NJ, Wick BJ, Johnson MJ, Webber BR, Moriarity BS. Engineering memory T cells as a platform for long-term enzyme replacement therapy in lysosomal storage disorders. Mol Ther 2024; 32:3865-3878. [PMID: 39367605 PMCID: PMC11573576 DOI: 10.1016/j.ymthe.2024.09.033] [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: 04/15/2024] [Revised: 08/19/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024] Open
Abstract
Enzymopathy disorders are the result of missing or defective enzymes. Among these enzymopathies, mucopolysaccharidosis type I is a rare genetic lysosomal storage disorder caused by mutations in the gene encoding alpha-L-iduronidase (IDUA), which ultimately causes toxic buildup of glycosaminoglycans (GAGs). There is currently no cure and standard treatments provide insufficient relief to the skeletal structure and central nervous system (CNS). Human memory T (Tm) cells migrate throughout the body's tissues and can persist for years, making them an attractive approach for cellular-based, systemic enzyme replacement therapy. Here, we tested genetically engineered, IDUA-expressing Tm cells as a cellular therapy in an immunodeficient mouse model of MPS I. Our results demonstrate that a single dose of engineered Tm cells leads to detectable IDUA enzyme levels in the blood for up to 22 weeks and reduced urinary GAG excretion. Furthermore, engineered Tm cells take up residence in nearly all tested tissues, producing IDUA and leading to metabolic correction of GAG levels in the heart, lung, liver, spleen, kidney, bone marrow, and the CNS, although only minimal improved cognition was observed. Our study indicates that genetically engineered Tm cells hold great promise as a platform for cellular-based enzyme replacement therapy for the treatment of mucopolysaccharidosis type I and potentially many other enzymopathies and protein deficiencies.
Collapse
Affiliation(s)
- Evan W Kleinboehl
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kanut Laoharawee
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jacob D Jensen
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Joseph J Peterson
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nicholas J Slipek
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Bryce J Wick
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Matthew J Johnson
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Beau R Webber
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Branden S Moriarity
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
36
|
Gupta S, Martinov T, Thelen A, Sunahara M, Mureli S, Vazquez A, Gerdts J, Dandekar R, Cortese I, Fouassier C, Schanzer E, Urnov FD, Marson A, Shy BR, Greenberg PD, Wilson MR. Antigen-Specific T Cell Receptor Discovery for Treating Progressive Multifocal Leukoencephalopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621904. [PMID: 39574748 PMCID: PMC11580961 DOI: 10.1101/2024.11.04.621904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Background Progressive multifocal leukoencephalopathy (PML) is a frequently fatal disease of the central nervous system caused by JC virus (JCV). Survival is dependent on early diagnosis and ability to re-establish anti-viral T cell immunity. Adoptive transfer of polyomavirus-specific T cells has shown promise; however, there are no readily available HLA-matched anti-viral T cells to facilitate rapid treatment. Objective Identify epitopes of the JCV major capsid protein VP1 that elicit an immune response in the context of human leukocyte antigen allele A*02:01 (HLA-A2) and isolate cognate T cell receptors (TCRs) from healthy donors. Evaluate individual VP1-specific TCRs for their capacity to be expressed in T cells and clear JCV in vitro . Methods PBMCs from HLA-A2+ healthy donors were stimulated with peptide libraries tiled across the JCV VP1 protein. Multiple rounds of stimulation were performed to identify the antigens that induced the largest expansion and CD8 + T cell response (measured as INF γ , TNF α , CD137, and CD69 expression). High-affinity, antigen-specific CD8 + T cells were isolated based on intensity of tetramer binding for downstream single-cell TCR sequencing. Candidate TCRs were selected based on tetramer binding affinity and activation assays. Promising TCRs were introduced into the T cell genome via viral transduction for in vitro validation including peptide-pulsed K562 cells and astrocyte cells, and JCV-infected astrocytes. Results Four conserved JCV VP1 epitopes (amino acids 100-108, 251-259, 253-262, and 274-283) presented by HLA-A2 were identified. VP1(100-108) consistently elicited the highest level of IFN- γ production from multiple donors and this peptide is in a highly conserved region of VP1. We next identified fourteen high avidity TCRs specific for VP1(100-108). When virally transduced into primary human T cells, seven of these TCRs demonstrated specific binding to VP1(100-108):HLA-A2 tetramers, and four showed increased IFN- γ response when incubated with peptide. Primary CD8 + T cells expressing two of these TCRs cleared both HLA-A2 positive K562 cells and HLA-A2 positive SVG astrocyte cell line presenting exogenously added VP1 peptide at a range of E:T ratios. In addition, both TCR-transduced T cell populations effectively lysed JCV-infected astrocytes. Conclusions We identified JCV VP1 epitopes that are immunogenic in the context of HLA-A2 MHC-I, including epitopes that have not been previously described. The VP1(100-108) epitope was used to isolate HLA-A2-restricted TCRs. When cloned into primary human CD8 + T cells, these TCRs recognized VP1 (100-108)-presenting targets, and the transduced T cells conferred cytotoxic activity and eliminated K562 and astrocyte cells displaying the VP1(100-108) peptide and not sham peptide, as well as JCV-infected astrocytes. Taken together, these data suggest that JCV VP1-specific TCRs could be appealing therapeutics for HLA-A2+ individuals with PML in whom intrinsic T cell immunity cannot be rescued.
Collapse
|
37
|
Mittl K, Hayashi F, Dandekar R, Schubert RD, Gerdts J, Oshiro L, Loudermilk R, Greenfield A, Augusto DG, Ramesh A, Tran E, Koshal K, Kizer K, Dreux J, Cagalingan A, Schustek F, Flood L, Moore T, Kirkemo LL, Cooper T, Harms M, Gomez R, Sibener L, Cree BAC, Hauser SL, Hollenbach JA, Gee M, Wilson MR, Zamvil SS, Sabatino JJ. Antigen specificity of clonally-enriched CD8+ T cells in multiple sclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.07.611010. [PMID: 39282370 PMCID: PMC11398516 DOI: 10.1101/2024.09.07.611010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
CD8+ T cells are the dominant lymphocyte population in multiple sclerosis (MS) lesions where they are highly clonally expanded. The clonal identity, function, and antigen specificity of CD8+ T cells in MS are not well understood. Here we report a comprehensive single-cell RNA-seq and T cell receptor (TCR)-seq analysis of the cerebrospinal fluid (CSF) and blood from a cohort of treatment-naïve MS patients and control participants. A small subset of highly expanded and activated CD8+ T cells were enriched in the CSF in MS that displayed high activation, cytotoxicity and tissue-homing transcriptional profiles. Using a combination of unbiased and targeted antigen discovery approaches, MS-derived CD8+ T cell clonotypes recognizing Epstein-Barr virus (EBV) antigens and multiple novel mimotopes were identified. These findings shed vital insight into the role of CD8+ T cells in MS and pave the way towards disease biomarkers and therapeutic targets.
Collapse
|
38
|
Yue T, Chen SY, Shen WK, Zhang ZY, Cheng L, Guo AY. TCRosetta: An Integrated Analysis and Annotation Platform for T-cell Receptor Sequences. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae013. [PMID: 39436242 PMCID: PMC11849489 DOI: 10.1093/gpbjnl/qzae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 12/23/2023] [Accepted: 01/08/2024] [Indexed: 10/23/2024]
Abstract
T cells and T-cell receptors (TCRs) are essential components of the adaptive immune system. Characterization of the TCR repertoire offers a promising and highly informative source for understanding the functions of T cells in the immune response and immunotherapy. Although TCR repertoire studies have attracted much attention, there are few online servers available for TCR repertoire analysis, especially for TCR sequence annotation or advanced analyses. Therefore, we developed TCRosetta, a comprehensive online server that integrates analytical methods for TCR repertoire analysis and visualization. TCRosetta combines general feature analysis, large-scale sequence clustering, network construction, peptide-TCR binding prediction, generation probability calculation, and k-mer motif analysis for TCR sequences, making TCR data analysis as simple as possible. The TCRosetta server accepts multiple input data formats and can analyze ∼ 20,000 TCR sequences in less than 3 min. TCRosetta is the most comprehensive web server available for TCR repertoire analysis and is freely available at https://guolab.wchscu.cn/TCRosetta/.
Collapse
Affiliation(s)
- Tao Yue
- Center for Artificial Intelligence Biology, Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Si-Yi Chen
- Center for Artificial Intelligence Biology, Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Wen-Kang Shen
- Center for Artificial Intelligence Biology, Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhan-Ye Zhang
- Center for Artificial Intelligence Biology, Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Liming Cheng
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - An-Yuan Guo
- Center for Artificial Intelligence Biology, Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
39
|
Yao ZQ, Schank MB, Zhao J, El Gazzar M, Wang L, Zhang Y, Hill AC, Banik P, Pyburn JS, Moorman JP. The potential of HBV cure: an overview of CRISPR-mediated HBV gene disruption. Front Genome Ed 2024; 6:1467449. [PMID: 39444780 PMCID: PMC11496132 DOI: 10.3389/fgeed.2024.1467449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024] Open
Abstract
Hepatitis B virus (HBV) infection is a common cause of liver disease worldwide. The current antiviral treatment using nucleotide analogues (NAs) can only suppress de novo HBV replication but cannot eliminate chronic HBV infection due to the persistence of covalently closed circular (ccc) DNA that sustains viral replication. The CRISPR/Cas9 system is a novel genome-editing tool that enables precise gene disruption and inactivation. With high efficiency and simplicity, the CRISPR/Cas9 system has been utilized in multiple studies to disrupt the HBV genome specifically, eliciting varying anti-HBV effects both in vitro and in vivo. Additionally, multi-locus gene targeting has shown enhanced antiviral activity, paving the way for combination therapy to disrupt and inactivate HBV cccDNA as well as integrated HBV DNA. Despite its promising antiviral effects, this technology faces several challenges that need to be overcome before its clinical application, i.e., off-target effects and in vivo drug delivery. As such, there is a need for improvement in CRISPR/Cas9 efficiency, specificity, versatility, and delivery. Here, we critically review the recent literature describing the tools employed in designing guide RNAs (gRNAs) targeting HBV genomes, the vehicles used for expressing and delivering CRISPR/Cas9 components, the models used for evaluating CRISPR-mediated HBV gene disruption, the methods used for assessing antiviral and off-target effects induced by CRISPR/Cas9-mediated HBV gene disruption, and the prospects of future directions and challenges in leveraging this HBV gene-editing approach, to advance the HBV treatment toward a clinical cure.
Collapse
Affiliation(s)
- Zhi Q. Yao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN, United States
- Hepatitis (HBV/HCV/HIV) Program, James H. Quillen VA Medical Center, Department of Veterans Affairs, Johnson City, TN, United States
| | - Madison B. Schank
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN, United States
| | - Juan Zhao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN, United States
| | - Mohamed El Gazzar
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN, United States
| | - Ling Wang
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN, United States
| | - Yi Zhang
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN, United States
| | - Addison C. Hill
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN, United States
| | - Puja Banik
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN, United States
| | - Jaeden S. Pyburn
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN, United States
| | - Jonathan P. Moorman
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN, United States
- Hepatitis (HBV/HCV/HIV) Program, James H. Quillen VA Medical Center, Department of Veterans Affairs, Johnson City, TN, United States
| |
Collapse
|
40
|
Hajiaghayi M, Gholizadeh F, Han E, Little SR, Rahbari N, Ardila I, Lopez Naranjo C, Tehranimeh K, Shih SCC, Darlington PJ. The β 2-adrenergic biased agonist nebivolol inhibits the development of Th17 and the response of memory Th17 cells in an NF-κB-dependent manner. Front Immunol 2024; 15:1446424. [PMID: 39445009 PMCID: PMC11496295 DOI: 10.3389/fimmu.2024.1446424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 09/11/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction Adrenergic receptors regulate metabolic, cardiovascular, and immunological functions in response to the sympathetic nervous system. The effect of β2-adrenergic receptor (AR) as a high expression receptor on different subpopulations of T cells is complex and varies depending on the type of ligand and context. While traditional β2-AR agonists generally suppress T cells, they potentially enhance IL-17A production by Th17 cells. The effects of pharmacological drugs that count as biased agonists of AR like nebivolol are not completely understood. We investigated the impact of nebivolol on human memory CD4+ T (Th1, Th2, Th17) cells and polarized naive Th17 cells, highlighting its potential for IL-17A suppression via a non-canonical β2-AR cell signaling pathway. Methods The effects of nebivolol were tested on healthy human peripheral blood mononuclear cells, purified memory Th cells, and polarized naive Th17 cells activated with anti-CD3/anti-CD28/anti-CD2 ImmunoCult reagent. IFN-γ, IL-4, and IL-17A, which are primarily derived from Th1, Th2, and Th17 cells, respectively, were quantified by ELISA and flow cytometry. IL-10 was measured by ELISA. Gene expression of RORC, ADRB1, ADRB2, and ADRB3 was evaluated by qPCR. The ADRB2 gene was knocked out in memory Th cells using CRISPR/Cas9. Protein expression of phosphorylated serine133-CREB and phosphorylated NF-κB p65 was assessed by Western blot. Proliferation was assessed by fluorescent dye loading and flow cytometry. Results Nebivolol treatment decreased IL-17A and IFN-γ secretion by activated memory Th cells and elevated IL-4 levels. Nebivolol reduced the proportion of IL-17A+ Th cells and downregulated RORC expression. Unlike the β2-AR agonist terbutaline, nebivolol inhibited the shift of naive CD4+ T cells toward the Th17 phenotype. IL-10 and the proliferation index remained unchanged. Nebivolol-treated β2-knockout memory Th cells showed significant inhibition of β2-AR-mediated signaling, evidenced by the absence of IL-17A suppression compared to controls. Phosphorylation of the NF-κB p65 subunit was inhibited by nebivolol, but CREB phosphorylation was not changed, suggesting a selective transcriptional control. Conclusions The findings demonstrate that nebivolol acts through a β2-AR-mediated signaling pathway, as a distinctive anti-inflammatory agent capable of selectively shifting Th17 cells and suppressing the phosphorylation of NF-κB. This highlights nebivolol's potential for therapeutic interventions in chronic autoimmune conditions with elevated IL-17A levels.
Collapse
Affiliation(s)
- Mehri Hajiaghayi
- Department of Biology, Concordia University, Montréal, QC, Canada
| | | | - Eric Han
- Department of Health, Kinesiology and Applied Physiology, Concordia University, Montréal QC, Canada
| | - Samuel R. Little
- Department of Electrical and Computer Engineering, Concordia University, Center of Applied Synthetic Biology, Montréal, QC, Canada
| | - Niloufar Rahbari
- Department of Electrical and Computer Engineering, Concordia University, Center of Applied Synthetic Biology, Montréal, QC, Canada
| | - Isabella Ardila
- Department of Biology, Concordia University, Montréal, QC, Canada
| | | | - Kasra Tehranimeh
- Department of Health, Kinesiology and Applied Physiology, Concordia University, Montréal QC, Canada
| | - Steve C. C. Shih
- Department of Electrical and Computer Engineering, Concordia University, Center of Applied Synthetic Biology, Montréal, QC, Canada
| | - Peter J. Darlington
- Department of Biology, Concordia University, Montréal, QC, Canada
- Department of Health, Kinesiology and Applied Physiology, Concordia University, Montréal QC, Canada
| |
Collapse
|
41
|
Chen X, Gao Y, Zhang Y. Allogeneic CAR-T cells for cancer immunotherapy. Immunotherapy 2024; 16:1079-1090. [PMID: 39378059 PMCID: PMC11492692 DOI: 10.1080/1750743x.2024.2408048] [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: 06/17/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
Autologous chimeric antigen receptor (CAR)-modified T (CAR-T) cell therapy has displayed high efficacy in the treatment of hematological malignancies. Up to now, 11 autologous CAR-T cell products have been approved for the management of malignancies globally. However, the application of autologous CAR-T cell therapy has many individual limitations, long time-consuming, highly cost, and the risk of manufacturing failure. Indeed, some patients would not benefit from autologous CAR-T cell products because of rapid disease progression. Allogeneic CAR-T cells especially universal CAR-T (U-CAR-T) cell therapy are superior to these challenges of autologous CAR-T cells. In this review, we describe basic study and clinical trials of U-CAR-T cell therapeutic methods for malignancies. In addition, we summarize the problems encountered and potential solutions.
Collapse
Affiliation(s)
- Xinfeng Chen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yaoxin Gao
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, Henan, 450052, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450052, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450052, China
| |
Collapse
|
42
|
Huang T, Bei C, Hu Z, Li Y. CAR-macrophage: Breaking new ground in cellular immunotherapy. Front Cell Dev Biol 2024; 12:1464218. [PMID: 39421021 PMCID: PMC11484238 DOI: 10.3389/fcell.2024.1464218] [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: 07/16/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Chimeric Antigen Receptor (CAR) technology has revolutionized cellular immunotherapy, particularly with the success of CAR-T cells in treating hematologic malignancies. However, CAR-T cells have the limited efficacy of against solid tumors. To address these limitations, CAR-macrophages (CAR-Ms) leverage the innate properties of macrophages with the specificity and potency of CAR technology, offering a novel and promising approach to cancer immunotherapy. Preclinical studies have shown that CAR-Ms can effectively target and destroy tumor cells, even within challenging microenvironments, by exhibiting direct cytotoxicity and enhancing the recruitment and activation of other immune cells. Additionally, the favorable safety profile of macrophages and their persistence within solid tumors position CAR-Ms as potentially safer and more durable therapeutic options compared to CAR-T cells. This review explores recent advancements in CAR-Ms technology, including engineering strategies to optimize their anti-tumor efficacy and preclinical evidence supporting their use. We also discuss the challenges and future directions in developing CAR-Ms therapies, emphasizing their potential to revolutionize cellular immunotherapy. By harnessing the unique properties of macrophages, CAR-Ms offer a groundbreaking approach to overcoming the current limitations of CAR-T cell therapies, paving the way for more effective and sustainable cancer treatments.
Collapse
Affiliation(s)
- Ting Huang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chenqi Bei
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhenhua Hu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
| | - Yuanyuan Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
43
|
Wang F, Huang Y, Li J, Zhou W, Wang W. Targeted gene delivery systems for T-cell engineering. Cell Oncol (Dordr) 2024; 47:1537-1560. [PMID: 38753155 DOI: 10.1007/s13402-024-00954-6] [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] [Accepted: 04/28/2024] [Indexed: 06/27/2024] Open
Abstract
T lymphocytes are indispensable for the host systems of defense against pathogens, tumors, and environmental threats. The therapeutic potential of harnessing the cytotoxic properties of T lymphocytes for antigen-specific cell elimination is both evident and efficacious. Genetically engineered T-cells, such as those employed in CAR-T and TCR-T cell therapies, have demonstrated significant clinical benefits in treating cancer and autoimmune disorders. However, the current landscape of T-cell genetic engineering is dominated by strategies that necessitate in vitro T-cell isolation and modification, which introduce complexity and prolong the development timeline of T-cell based immunotherapies. This review explores the complexities of gene delivery systems designed for T cells, covering both viral and nonviral vectors. Viral vectors are known for their high transduction efficiency, yet they face significant limitations, such as potential immunogenicity and the complexities involved in large-scale production. Nonviral vectors, conversely, offer a safer profile and the potential for scalable manufacturing, yet they often struggle with lower transduction efficiency. The pursuit of gene delivery systems that can achieve targeted gene transfer to T cell without the need for isolation represents a significant advancement in the field. This review assesses the design principles and current research progress of such systems, highlighting the potential for in vivo gene modification therapies that could revolutionize T-cell based treatments. By providing a comprehensive analysis of these systems, we aim to contribute valuable insights into the future development of T-cell immunotherapy.
Collapse
Affiliation(s)
- Fengling Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Yong Huang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - JiaQian Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Weilin Zhou
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Wei Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
44
|
George CA, Sahu SU, de Oñate L, Souza BSDF, Wilson RC. Genome Editing Therapy for the Blood: Ex Vivo Success and In Vivo Prospects. CRISPR J 2024; 7:231-248. [PMID: 39324895 DOI: 10.1089/crispr.2024.0036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024] Open
Abstract
Hematopoietic stem cells (HSCs) provide the body with a continuous supply of healthy, functional blood cells. In patients with hematopoietic malignancies, immunodeficiencies, lysosomal storage disorders, and hemoglobinopathies, therapeutic genome editing offers hope for corrective intervention, with even modest editing efficiencies likely to provide clinical benefit. Engineered white blood cells, such as T cells, can be applied therapeutically to address monogenic disorders of the immune system, HIV infection, or cancer. The versatility of CRISPR-based tools allows countless new medical interventions for diseases of the blood, and rapid ex vivo success has been demonstrated in hemoglobinopathies via transplantation of the patient's HSCs following genome editing in a laboratory setting. Here we review recent advances in therapeutic genome editing of HSCs and T cells, focusing on the progress in ex vivo contexts, the promise of improved access via in vivo delivery, as well as the ongoing preclinical efforts that may enable the transition from ex vivo to in vivo administration. We discuss the challenges, limitations, and future prospects of this rapidly developing field, which may one day establish CRISPR as the standard of care for some diseases affecting the blood.
Collapse
Affiliation(s)
- Christy A George
- Innovative Genomics Institute, University of California Berkeley, Berkeley, California, USA
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California, USA
| | - Srishti U Sahu
- Innovative Genomics Institute, University of California Berkeley, Berkeley, California, USA
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California, USA
| | - Lorena de Oñate
- Innovative Genomics Institute, University of California Berkeley, Berkeley, California, USA
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California, USA
| | - Bruno Solano de Freitas Souza
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- Pioneer Science Initiative, D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Ross C Wilson
- Innovative Genomics Institute, University of California Berkeley, Berkeley, California, USA
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California, USA
- California Institute for Quantitative Biosciences, University of California Berkeley, Berkeley, California, USA
| |
Collapse
|
45
|
Lu ZX, Khimani AH, Tong Y. Importance of a reference ladder in analytical testing of CRISPR HDR donor ssDNA templates. Biotechniques 2024; 76:479-484. [PMID: 39632511 DOI: 10.1080/07366205.2024.2429308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024] Open
|
46
|
Yao Z, Zeng Y, Liu C, Jin H, Wang H, Zhang Y, Ding C, Chen G, Wu D. Focusing on CD8 + T-cell phenotypes: improving solid tumor therapy. J Exp Clin Cancer Res 2024; 43:266. [PMID: 39342365 PMCID: PMC11437975 DOI: 10.1186/s13046-024-03195-5] [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: 07/31/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Vigorous CD8+ T cells play a crucial role in recognizing tumor cells and combating solid tumors. How T cells efficiently recognize and target tumor antigens, and how they maintain the activity in the "rejection" of solid tumor microenvironment, are major concerns. Recent advances in understanding of the immunological trajectory and lifespan of CD8+ T cells have provided guidance for the design of more optimal anti-tumor immunotherapy regimens. Here, we review the newly discovered methods to enhance the function of CD8+ T cells against solid tumors, focusing on optimizing T cell receptor (TCR) expression, improving antigen recognition by engineered T cells, enhancing signal transduction of the TCR-CD3 complex, inducing the homing of polyclonal functional T cells to tumors, reversing T cell exhaustion under chronic antigen stimulation, and reprogramming the energy and metabolic pathways of T cells. We also discuss how to participate in the epigenetic changes of CD8+ T cells to regulate two key indicators of anti-tumor responses, namely effectiveness and persistence.
Collapse
Affiliation(s)
- Zhouchi Yao
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yayun Zeng
- Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Cheng Liu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Huimin Jin
- Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Hong Wang
- Department of Scientific Research, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Yue Zhang
- Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Chengming Ding
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Guodong Chen
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Daichao Wu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
- Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
47
|
Kattelus R, Starskaia I, Lindén M, Batkulwar K, Pietilä S, Moulder R, Marson A, Rasool O, Suomi T, Elo LL, Lahesmaa R, Buchacher T. Phenotypic profiling of human induced regulatory T cells at early differentiation: insights into distinct immunosuppressive potential. Cell Mol Life Sci 2024; 81:399. [PMID: 39264416 PMCID: PMC11393232 DOI: 10.1007/s00018-024-05429-3] [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: 06/26/2024] [Revised: 08/20/2024] [Accepted: 08/27/2024] [Indexed: 09/13/2024]
Abstract
Regulatory T cells (Tregs) play a key role in suppressing systemic effector immune responses, thereby preventing autoimmune diseases but also potentially contributing to tumor progression. Thus, there is great interest in clinically manipulating Tregs, but the precise mechanisms governing in vitro-induced Treg (iTreg) differentiation are not yet fully understood. Here, we used multiparametric mass cytometry to phenotypically profile human iTregs during the early stages of in vitro differentiation at single-cell level. A panel of 25 metal-conjugated antibodies specific to markers associated with human Tregs was used to characterize these immunomodulatory cells. We found that iTregs highly express the transcription factor FOXP3, as well as characteristic Treg-associated surface markers (e.g. CD25, PD1, CD137, CCR4, CCR7, CXCR3, and CD103). Expression of co-inhibitory factors (e.g. TIM3, LAG3, and TIGIT) increased slightly at late stages of iTreg differentiation. Further, CD103 was upregulated on a subpopulation of iTregs with greater suppressive capacity than their CD103- counterparts. Using mass-spectrometry-based proteomics, we showed that sorted CD103+ iTregs express factors associated with immunosuppression. Overall, our study highlights that during early stages of differentiation, iTregs resemble memory-like Treg features with immunosuppressive activity, and provides opportunities for further investigation into the molecular mechanisms underlying Treg function.
Collapse
Affiliation(s)
- Roosa Kattelus
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Turku Doctoral Programme of Molecular Medicine, University of Turku, Turku, Finland
| | - Inna Starskaia
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Turku Doctoral Programme of Molecular Medicine, University of Turku, Turku, Finland
| | - Markus Lindén
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Kedar Batkulwar
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Sami Pietilä
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Robert Moulder
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Alexander Marson
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, 94158, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Omid Rasool
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Tomi Suomi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Laura L Elo
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Riitta Lahesmaa
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
- Institute of Biomedicine, University of Turku, Turku, Finland.
| | - Tanja Buchacher
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
| |
Collapse
|
48
|
Peng D, Vangipuram M, Wong J, Leonetti M. protoSpaceJAM: an open-source, customizable and web-accessible design platform for CRISPR/Cas insertional knock-in. Nucleic Acids Res 2024; 52:e68. [PMID: 38922690 PMCID: PMC11347160 DOI: 10.1093/nar/gkae553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
CRISPR/Cas-mediated knock-in of DNA sequences enables precise genome engineering for research and therapeutic applications. However, designing effective guide RNAs (gRNAs) and homology-directed repair (HDR) donors remains a bottleneck. Here, we present protoSpaceJAM, an open-source algorithm to automate and optimize gRNA and HDR donor design for CRISPR/Cas insertional knock-in experiments, currently supporting SpCas9, SpCas9-VQR and enAsCas12a Cas enzymes. protoSpaceJAM utilizes biological rules to rank gRNAs based on specificity, distance to insertion site, and position relative to regulatory regions. protoSpaceJAM can introduce 'recoding' mutations (silent mutations and mutations in non-coding sequences) in HDR donors to prevent re-cutting and increase knock-in efficiency. Users can customize parameters and design double-stranded or single-stranded donors. We validated protoSpaceJAM's design rules by demonstrating increased knock-in efficiency with recoding mutations and optimal strand selection for single-stranded donors. An additional module enables the design of genotyping primers for deep sequencing of edited alleles. Overall, protoSpaceJAM streamlines and optimizes CRISPR knock-in experimental design in a flexible and modular manner to benefit diverse research and therapeutic applications. protoSpaceJAM is available open-source as an interactive web tool at protospacejam.czbiohub.org or as a standalone Python package at github.com/czbiohub-sf/protoSpaceJAM.
Collapse
Affiliation(s)
- Duo Peng
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | | | - Joan Wong
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | | |
Collapse
|
49
|
Lu J, Ma Y, Li Q, Xu Y, Xue Y, Xu S. CAR Macrophages: a promising novel immunotherapy for solid tumors and beyond. Biomark Res 2024; 12:86. [PMID: 39175095 PMCID: PMC11342599 DOI: 10.1186/s40364-024-00637-2] [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: 07/08/2024] [Accepted: 08/09/2024] [Indexed: 08/24/2024] Open
Abstract
With the advent of adoptive cellular therapy, chimeric antigen receptor (CAR)-T cell therapy has gained widespread application in cancer treatment and has demonstrated significant efficacy against certain hematologic malignancies. However, due to the limitations of CAR-T cell therapy in treating solid tumors, other immune cells are being modified with CAR to address this issue. Macrophages have emerged as a promising option, owing to their extensive immune functions, which include antigen presentation, powerful tumor phagocytosis, and particularly active trafficking to the tumor microenvironment. Leveraging their unique advantages, CAR-macrophages (CAR-M) are expected to enhance the effectiveness of solid tumor treatments as a novel form of immunotherapy, potentially overcoming major challenges associated with CAR-T/NK therapy. This review outlines the primary mechanism underlying CAR-M and recent progressions in CAR-M therapy, while also discussing their further applications.
Collapse
Affiliation(s)
- Jialin Lu
- National Key Lab of Immunity and Inflammation and Institute of Immunology, Naval Medical University/Second Military Medical University, Shanghai, 200433, China
| | - Yuqing Ma
- National Key Lab of Immunity and Inflammation and Institute of Immunology, Naval Medical University/Second Military Medical University, Shanghai, 200433, China
| | - Qiuxin Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yihuan Xu
- National Key Lab of Immunity and Inflammation and Institute of Immunology, Naval Medical University/Second Military Medical University, Shanghai, 200433, China
| | - Yiquan Xue
- National Key Lab of Immunity and Inflammation and Institute of Immunology, Naval Medical University/Second Military Medical University, Shanghai, 200433, China.
| | - Sheng Xu
- National Key Lab of Immunity and Inflammation and Institute of Immunology, Naval Medical University/Second Military Medical University, Shanghai, 200433, China.
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China.
| |
Collapse
|
50
|
Moshref M, Lo JHH, McKay A, Camperi J, Schroer J, Ueno N, Wang S, Gulati S, Tarighat S, Durinck S, Lee HY, Chen D. Assessing a single-cell multi-omic analytic platform to characterize ex vivo-engineered T-cell therapy products. Front Bioeng Biotechnol 2024; 12:1417070. [PMID: 39229457 PMCID: PMC11368872 DOI: 10.3389/fbioe.2024.1417070] [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: 04/13/2024] [Accepted: 07/11/2024] [Indexed: 09/05/2024] Open
Abstract
Genetically engineered CD8+ T cells are being explored for the treatment of various cancers. Analytical characterization represents a major challenge in the development of genetically engineered cell therapies, especially assessing the potential off-target editing and product heterogeneity. As conventional sequencing techniques only provide information at the bulk level, they are unable to detect off-target CRISPR translocation or editing events occurring in minor cell subpopulations. In this study, we report the analytical development of a single-cell multi-omics DNA and protein assay to characterize genetically engineered cell products for safety and genotoxicity assessment. We were able to quantify on-target edits, off-target events, and potential translocations at the targeting loci with per-cell granularity, providing important characterization data of the final cell product. Conclusion: A single-cell multi-omics approach provides the resolution required to understand the composition of cellular products and identify critical quality attributes (CQAs).
Collapse
Affiliation(s)
- Maryam Moshref
- Cell Therapy Engineering and Development, Genentech, South San Francisco, CA, United States
| | - Jerry Hung-Hao Lo
- Oncology Bioinformatics, Genentech, South San Francisco, CA, United States
| | - Andrew McKay
- Pharma Technical Development Bioinformatics, Genentech, South San Francisco, CA, United States
| | - Julien Camperi
- Cell Therapy Engineering and Development, Genentech, South San Francisco, CA, United States
| | - Joseph Schroer
- Cell Therapy Engineering and Development, Genentech, South San Francisco, CA, United States
| | - Norikiyo Ueno
- Cell and Gene Therapy Business Unit, Mission Bio, South San Francisco, CA, United States
| | - Shu Wang
- Bioinformatics Department, Mission Bio, South San Francisco, CA, United States
| | - Saurabh Gulati
- Bioinformatics Department, Mission Bio, South San Francisco, CA, United States
| | - Somayeh Tarighat
- Cell Therapy Engineering and Development, Genentech, South San Francisco, CA, United States
| | - Steffen Durinck
- Oncology Bioinformatics, Genentech, South San Francisco, CA, United States
| | - Ho Young Lee
- Cell Therapy Engineering and Development, Genentech, South San Francisco, CA, United States
| | - Dayue Chen
- Cell Therapy Engineering and Development, Genentech, South San Francisco, CA, United States
| |
Collapse
|