1
|
Xu J, Jia Z, Zhao X, Wang L, Jin G, Li Z, Yin N, Li Y, Peng M. BCOR and ZC3H12A suppress a core stemness program in exhausted CD8+ T cells. J Exp Med 2025; 222:e20241133. [PMID: 40327039 PMCID: PMC12054362 DOI: 10.1084/jem.20241133] [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: 07/02/2024] [Revised: 11/25/2024] [Accepted: 04/16/2025] [Indexed: 05/07/2025] Open
Abstract
In chronic viral infections, sustained CD8+ T cell response relies on TCF1+ precursor-exhausted T cells (TPEX) exhibiting stem-like properties. TPEX self-renew and respond to PD-1 blockade, underscoring their paramount importance. However, strategies for effectively augmenting TPEX remain limited. Here, we demonstrate that ZC3H12A deficiency initiates a stemness program in TPEX but also increases cell death, whereas BCOR deficiency predominantly promotes TPEX proliferation. Consequently, co-targeting of both BCOR and ZC3H12A imparts exceptional stemness and functionality to TPEX, thereby enhancing viral control. Mechanistically, BCOR and ZC3H12A collaboratively suppress a core stemness program in TPEX characterized by heightened expression of ∼216 factors. While TCF1 plays a role, this core stemness program relies on novel factors, including PDZK1IP1, IFIT3, PIM2, LTB, and POU2F2. Crucially, overexpressing POU2F2 robustly boosts TPEX and enhances antiviral immunity. Thus, a core stemness program exists in exhausted T cells, jointly repressed by BCOR and ZC3H12A, robustly controlling TPEX differentiation and providing new targets for addressing T cell exhaustion.
Collapse
Affiliation(s)
- Jing Xu
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zeran Jia
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- IDG/McGovern Institute for Brain Research, State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Xiaocui Zhao
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Lixia Wang
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Gang Jin
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zhuoyang Li
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Na Yin
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Yinqing Li
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- IDG/McGovern Institute for Brain Research, State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Min Peng
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| |
Collapse
|
2
|
Green WD, Gomez A, Plotkin AL, Pratt BM, Merritt EF, Mullins GN, Kren NP, Modliszewski JL, Zhabotynsky V, Woodcock MG, Green JM, Cannon G, Pipkin ME, Dotti G, Thaxton JE, Pylayeva-Gupta Y, Baldwin AS, Morris JP, Stanley N, Milner JJ. Enhancer-driven gene regulatory networks reveal transcription factors governing T cell adaptation and differentiation in the tumor microenvironment. Immunity 2025:S1074-7613(25)00193-1. [PMID: 40425012 DOI: 10.1016/j.immuni.2025.04.030] [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: 03/12/2024] [Revised: 02/11/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025]
Abstract
Tumor-infiltrating lymphocytes (TILs) with a tissue-resident memory CD8+ T cell (Trm) phenotype are associated with improved patient outcomes in solid malignancies. To define programs governing the formation of Trm-like TIL, we performed paired single-cell RNA sequencing and single-cell ATAC sequencing of T cell receptor (TCR)-matched CD8+ T cells in models of infection and cancer. Enhancer-driven regulons assembled from multiomic profiling data revealed epigenetic and transcriptional programs regulating the formation of Trm-like TIL in relation to canonical exhausted and memory T cell states. The transcriptional regulator KLF2 repressed the formation of CD69+CD103+ Trm-like TIL and limited anti-tumor activity. Conversely, sustained expression of the transcription factor BATF enhanced formation of CD69+CD103+ TIL, contingent upon downregulation of KLF2. Transforming growth factor β (TGF-β) signaling and CD103 expression were necessary for Trm-like TIL formation, but BATF overexpression was sufficient to drive formation of CD69+CD103+ TIL in TGFBR2-silenced cells. These findings reveal mechanisms of Trm-like TIL differentiation and provide a framework for considering tissue residency in the context of CD8+ T cell heterogeneity in the tumor microenvironment.
Collapse
Affiliation(s)
- William D Green
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Amber Gomez
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alec L Plotkin
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Computational Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27559, USA
| | - Brandon M Pratt
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Emily F Merritt
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Genevieve N Mullins
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nancy P Kren
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27559, USA
| | - Jennifer L Modliszewski
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Vasyl Zhabotynsky
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mark G Woodcock
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jarred M Green
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Gabrielle Cannon
- Advanced Analytics Core, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27559, USA
| | - Matthew E Pipkin
- Department of Immunology and Microbiology, UF Scripps Biomedical Research, University of Florida, Jupiter, FL 33458, USA
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jessica E Thaxton
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yuliya Pylayeva-Gupta
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27559, USA
| | - Albert S Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - John P Morris
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Natalie Stanley
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Computational Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - J Justin Milner
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
3
|
Zhu Z, Lou G, Luo Y, Yihunie K, Hoar J, Daniel JA, Evers BM, Yao C, Wu T. Aging Compromises Terminal Differentiation Program of Cytotoxic Effector Lineage and Promotes Exhaustion in CD8 + T Cells Responding to Coronavirus Infection. Aging Cell 2025:e70109. [PMID: 40396260 DOI: 10.1111/acel.70109] [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: 12/26/2024] [Revised: 04/16/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025] Open
Abstract
T cell aging increases the risk of viral infection-related morbidity and mortality and reduces vaccine efficacy in the elderly. A major hallmark of T cell aging is the loss of quiescence and shift toward terminal differentiation during homeostasis. However, how aging impacts the differentiation program of virus-specific T cells during infection is unclear. Here, in a murine coronavirus (MHV) infection model with age-associated increased mortality, we demonstrate that aging impairs, instead of promoting, the terminal differentiation program of virus-specific CD8+ T cells. Upon infection, CD8+ and CD4+ T cells in old mice showed marked reduction in clonal expansion and upregulation of immune checkpoints associated with T cell exhaustion. Bulk and single-cell transcriptomics showed that aging upregulated the T cell exhaustion transcriptional program associated with TOX in virus-specific CD8+ T cells and shifted the myeloid compartment from immunostimulatory to immunosuppressive phenotype. In addition, aging downregulated the transcriptional program of terminally differentiated effector CD8+ T cells and diminished the CX3CR1+ cytotoxic effector lineage. Mechanistically, virus-specific CD8+ T cells from infected aged mice displayed defects in inducing transcription factors ZEB2 and KLF2, which were required for terminal differentiation of effector CD8+ T cells. Together, our study shows that aging impairs terminal differentiation and promotes exhaustion of virus-specific CD8+ T cells responding to coronavirus infection through dysregulating expression of lineage-defining transcription factors.
Collapse
Affiliation(s)
- Ziang Zhu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Immunology Ph.D. Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Guohua Lou
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ying Luo
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kiddist Yihunie
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cancer Biology Ph.D. Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jonathan Hoar
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Juan A Daniel
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bret M Evers
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chen Yao
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tuoqi Wu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
4
|
Mahyari E, Boggy GJ, McElfresh GW, Kaza M, Benjamin S, Varco-Merth B, Ojha S, Feltham S, Goodwin W, Nkoy C, Duell D, Selseth A, Bennett T, Barber-Axthelm A, Smedley JV, Labriola CS, Axthelm MK, Reeves RK, Okoye AA, Hansen SG, Picker LJ, Bimber BN. Enhanced interpretation of immune cell phenotype and function through a rhesus macaque single-cell atlas. CELL GENOMICS 2025; 5:100849. [PMID: 40233746 DOI: 10.1016/j.xgen.2025.100849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/17/2025] [Accepted: 03/18/2025] [Indexed: 04/17/2025]
Abstract
Single-cell RNA sequencing (scRNA-seq) allows cell classification using genome-wide transcriptional state; however, high-dimensional transcriptomic profiles, and the unsupervised analyses employed to interpret them, provide a systematically different view of biology than well-established functional/lineage definitions of immunocytes. Understanding these differences and limits is essential for accurate interpretation of these rich data. We present the Rhesus Immune Reference Atlas (RIRA), the first immune-focused macaque single-cell multi-tissue atlas. We contrasted transcriptional profiles against immune lineages, using surface protein and marker genes as ground truth. While the pattern of clustering can align with cell type, this is not always true. Especially within T and natural killer (NK) cells, many functionally distinct subsets lack defining markers, and strong shared expression programs, such as cytotoxicity, result in systematic intermingling by unsupervised clustering. We identified gene programs with high discriminatory/diagnostic value, including multi-gene signatures that model T/NK cell maturation. Directly measuring these diagnostic programs complements unsupervised analyses.
Collapse
Affiliation(s)
- Eisa Mahyari
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Gregory J Boggy
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - G W McElfresh
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Maanasa Kaza
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Sebastian Benjamin
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Benjamin Varco-Merth
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Sohita Ojha
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Shana Feltham
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - William Goodwin
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Candice Nkoy
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Derick Duell
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Andrea Selseth
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Tyler Bennett
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Aaron Barber-Axthelm
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Jeremy V Smedley
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Caralyn S Labriola
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Michael K Axthelm
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - R Keith Reeves
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Duke University School of Medicine, Durham, NC, USA; Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Afam A Okoye
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Scott G Hansen
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Louis J Picker
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Benjamin N Bimber
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA.
| |
Collapse
|
5
|
Hu W, Shawn Hu S, Zhu S, Peng W, Badovinac VP, Zang C, Zhao X, Xue HH. Hdac1 as an early determinant of intermediate-exhausted CD8 + T cell fate in chronic viral infection. Proc Natl Acad Sci U S A 2025; 122:e2502256122. [PMID: 40333757 DOI: 10.1073/pnas.2502256122] [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: 01/31/2025] [Accepted: 04/08/2025] [Indexed: 05/09/2025] Open
Abstract
The exhausted CD8+ T (TEX) cells consist of distinct subsets including Tcf1+ stem-like, Tcf1-Cx3cr1+ intermediate (TEX-int) and Tcf1-Cx3cr1- terminally exhausted cells; yet, epigenetic determinants of TEX subset differentiation remain incompletely understood. Using chronic viral infection, we show that histone deacetylase 1 (Hdac1) was specifically required for the formation of antigen-specific TEX-int cells at the effector phase of responses. Single-cell transcriptomics validated that Hdac1 deficiency depleted TEX-int cells and revealed that Hdac1 was critical for positive regulation of TEX-int-characteristic genes, including Cx3cr1, Cxcr6, and Klf2. Furthermore, profiling chromatin accessibility landscape in TEX subsets demonstrated that loss of Hdac1 resulted in a prevalent increase in chromatin open state, as evidently observed at the exhaustion program genes, which were linked to induced expression of exhaustion-inducing Tox transcription factor, PD1 and Lag3 coinhibitory receptors in TEX cells. Hdac1 thus has dual regulatory functions: promoting TEX-int cell fate and preventing excessive activation of the exhaustion program to curtail uncontrolled virus replication.
Collapse
Affiliation(s)
- Wei Hu
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ 07110
| | - Shengen Shawn Hu
- Department of Genome Sciences and University of Virginia Comprehensive Cancer Center, University of Virginia, Charlottesville, VA 22908
| | - Shaoqi Zhu
- Department of Physics, The George Washington University, Washington, DC 20052
- Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen 518107, Guangdong, China
| | - Weiqun Peng
- Department of Physics, The George Washington University, Washington, DC 20052
| | | | - Chongzhi Zang
- Department of Genome Sciences and University of Virginia Comprehensive Cancer Center, University of Virginia, Charlottesville, VA 22908
| | - Xudong Zhao
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Hai-Hui Xue
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ 07110
- Division of Research and Development, New Jersey Veterans Affairs Health Care System, East Orange, NJ 07018
| |
Collapse
|
6
|
Caballero AC, Ujaldón-Miró C, Pujol-Fernández P, Montserrat-Torres R, Guardiola-Perello M, Escudero-López E, Garcia-Cadenas I, Esquirol A, Martino R, Jara-Bustamante P, Ezquerra P, Soria JM, Iranzo E, Moreno-Martinez ME, Riba M, Sierra J, Alvarez-Fernández C, Escribà-Garcia L, Briones J. HSP-CAR30 with a high proportion of less-differentiated T cells promotes durable responses in refractory CD30+ lymphoma. Blood 2025; 145:1788-1801. [PMID: 39841453 DOI: 10.1182/blood.2024026758] [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: 09/03/2024] [Revised: 12/23/2024] [Accepted: 12/23/2024] [Indexed: 01/23/2025] Open
Abstract
ABSTRACT CD30-directed chimeric antigen receptor T-cell therapy (CART30) has limited efficacy in relapsed or refractory patients with CD30+ lymphoma, with a low proportion of durable responses. We have developed an academic CART30 cell product (HSP-CAR30) by combining strategies to improve performance. HSP-CAR30 targets a proximal epitope within the nonsoluble part of CD30, and the manufacturing process includes a modulation of ex vivo T-cell activation, as well as the addition of interleukin-21 (IL-21) to IL-7 and IL-15 to promote stemness of T cells. We translated HSP-CAR30 to a phase 1 clinical trial of 10 patients with relapsed/refractory classic Hodgkin lymphoma (HL) or CD30+ T-cell non-Hodgkin lymphoma. HSP-CAR30 was mainly composed of memory stem-like (TSCM-like) and central memory (TCM) CAR30+ T cells (87.5% ± 5%). No dose-limiting toxicities were detected. Six patients had grade 1 cytokine release syndrome, and no patient developed neurotoxicity. The overall response rate was 100%, and 5 of 8 patients with HL achieved complete remission (CR). An additional patient with HL achieved CR after a second HSP-CAR30 infusion. Remarkably, 60% of patients have ongoing CR after a mean follow-up of 34 months. CAR30+ T cells at expansion peak had a predominance of TSCM and TCM cells, and CAR30+ T cells remained detectable in 3 of 5 evaluable patients at least 12 months after infusion. Our study shows that selection of the epitope targeting CD30 and ex vivo preservation of less-differentiated memory T cells may enhance the efficacy of CART30 in patients with refractory HL. This trial is registered at www.clinicaltrials.gov (NCT04653649).
Collapse
Affiliation(s)
- Ana Carolina Caballero
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Cristina Ujaldón-Miró
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Paula Pujol-Fernández
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Rosanna Montserrat-Torres
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Maria Guardiola-Perello
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Eva Escudero-López
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | | | - Albert Esquirol
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Rodrigo Martino
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Paola Jara-Bustamante
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Pol Ezquerra
- Unit of Genomics of Complex Disease, Research Institute of Sant Pau Hospital, Barcelona, Spain
- Centre for Biomedical Network Research on Rare Diseases, Instituto de Salud Carlos III, Madrid, Spain
- Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
| | - José Manuel Soria
- Unit of Genomics of Complex Disease, Research Institute of Sant Pau Hospital, Barcelona, Spain
- Centre for Biomedical Network Research on Rare Diseases, Instituto de Salud Carlos III, Madrid, Spain
- Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
| | - Eva Iranzo
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Maria-Estela Moreno-Martinez
- Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Pharmacy Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Mireia Riba
- Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Pharmacy Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Jorge Sierra
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
| | - Carmen Alvarez-Fernández
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Laura Escribà-Garcia
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Javier Briones
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| |
Collapse
|
7
|
Jadlowsky JK, Hexner EO, Marshall A, Grupp SA, Frey NV, Riley JL, Veloso E, McConville H, Rogal W, Czuczman C, Hwang WT, Li Y, Leskowitz RM, Farrelly O, Karar J, Christensen S, Barber-Rotenberg J, Gaymon A, Aronson N, Bernstein W, Melenhorst JJ, Roche AM, Everett JK, Zolnoski SA, McFarland AG, Reddy S, Petrichenko A, Cook EJ, Lee C, Gonzalez VE, Alexander K, Kulikovskaya I, Ramírez-Fernández Á, Minehart JC, Ruella M, Gill SI, Schuster SJ, Cohen AD, Garfall AL, Shah PD, Porter DL, Maude SL, Levine BL, Siegel DL, Chew A, McKenna S, Lledo L, Davis MM, Plesa G, Herbst F, Stadtmauer EA, Tebas P, DiNofia A, Haas A, Haas NB, Myers R, O'Rourke DM, Svoboda J, Tanyi JL, Aplenc R, Jacobson JM, Ko AH, Cohen RB, June CH, Bushman FD, Fraietta JA. Long-term safety of lentiviral or gammaretroviral gene-modified T cell therapies. Nat Med 2025; 31:1134-1144. [PMID: 39833408 DOI: 10.1038/s41591-024-03478-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025]
Abstract
Long-term risks of gene therapy are not fully understood. In this study, we evaluated safety outcomes in 783 patients over more than 2,200 total patient-years of observation from 38 T cell therapy trials. The trials employed integrating gammaretroviral or lentiviral vectors to deliver engineered receptors to target HIV-1 infection or cancer. Eighteen patients (2.3%) developed secondary malignancies after treatment, with a median onset of 1.94 years (range: 51 d to 14 years). Where possible, incident tumor samples were analyzed for vector copy number, revealing no evidence of high-level marking or other indications of insertional mutagenesis. One T cell lymphoma was detected, but malignant T cells were not marked by vector integration. Analysis of vector integration sites in 176 patients revealed no pathological insertions linked to secondary malignancies, although, in some cases, integration in or near specific genes, including tumor suppressor genes, was associated with modest clonal expansion and sustained T cell persistence. These findings highlight the safety of engineered T cell therapies.
Collapse
Affiliation(s)
- Julie K Jadlowsky
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth O Hexner
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy Marshall
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephan A Grupp
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Noelle V Frey
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James L Riley
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Veloso
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Holly McConville
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Walter Rogal
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cory Czuczman
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei-Ting Hwang
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yimei Li
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel M Leskowitz
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Olivia Farrelly
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jayashree Karar
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shannon Christensen
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Julie Barber-Rotenberg
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Avery Gaymon
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Naomi Aronson
- Department of Medicine, Division of Infectious Diseases, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Wendy Bernstein
- Department of Medicine, Division of Infectious Diseases, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Jan Joseph Melenhorst
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Aoife M Roche
- 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
| | - Sonja A Zolnoski
- 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
| | - Shantan Reddy
- 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
| | - Emma J Cook
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carole Lee
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vanessa E Gonzalez
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kathleen Alexander
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Irina Kulikovskaya
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ángel Ramírez-Fernández
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Janna C Minehart
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA
| | - Saar I Gill
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen J Schuster
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adam D Cohen
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alfred L Garfall
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Payal D Shah
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David L Porter
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shannon L Maude
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Bruce L Levine
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Donald L Siegel
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anne Chew
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen McKenna
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lester Lledo
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Megan M Davis
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gabriela Plesa
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Friederike Herbst
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward A Stadtmauer
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Pablo Tebas
- Department of Medicine, Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amanda DiNofia
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Andrew Haas
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Naomi B Haas
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Regina Myers
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Donald M O'Rourke
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jakub Svoboda
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Janos L Tanyi
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard Aplenc
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jeffrey M Jacobson
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Andrew H Ko
- Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco, San Francisco, CA, USA
| | - Roger B Cohen
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, 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.
| | - Joseph A Fraietta
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Knox K, Jeltema D, Dobbs N, Yang K, Xing C, Song K, Tang Z, Torres-Ramirez G, Wang J, Gao S, Wu T, Yao C, Wang J, Yan N. Dynamic STING repression orchestrates immune cell development and function. Sci Immunol 2025; 10:eado9933. [PMID: 40053603 DOI: 10.1126/sciimmunol.ado9933] [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: 03/01/2024] [Revised: 11/15/2024] [Accepted: 01/17/2025] [Indexed: 03/09/2025]
Abstract
STING is an essential component of the innate immune system, yet homeostatic STING expression patterns and regulation are unknown. Using Sting1IRES-EGFP reporter and conditional Sting1 transgenic mice, we found that regulation of STING expression is critical for immune cell development and functionality. STING expression was repressed in neutrophils, and forced STING expression or signaling drove systemic inflammatory disease. During T lymphocyte development, STING expression was restricted at the double-positive stage via epigenetic silencing by DNA methyltransferase 1. Forced STING expression or signaling impaired T lymphocyte development independent of type I interferon and promoted lineage commitment to innate-like γδ T cells over adaptive αβ T cells. In the tumor microenvironment, CD8+ T lymphocytes repressed STING expression, correlating with features of T cell exhaustion in syngeneic mouse tumors and human colorectal cancer. Our data demonstrate the necessity of controlled, rather than ubiquitous, STING expression, uncovering a previously unappreciated dimension of STING pathobiology.
Collapse
Affiliation(s)
- Kennady Knox
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Devon Jeltema
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nicole Dobbs
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kun Yang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Cong Xing
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kun Song
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zhen Tang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gustavo Torres-Ramirez
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jiefu Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin 300060, China
| | - Shan Gao
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin 300060, China
| | - Tuoqi Wu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chen Yao
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jian Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin 300060, China
| | - Nan Yan
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
9
|
Qin D, Lei Y, Shu P, Zhang Y, Loh YH, Wang Y, Li Q. Supercharging CAR-T cells through transcriptional and epigenetic armoring. Theranostics 2025; 15:3345-3367. [PMID: 40093905 PMCID: PMC11905144 DOI: 10.7150/thno.107908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
Inspired by the remarkable success of CAR-T therapy in hematologic malignancies, research is increasingly focused on adapting this treatment for solid tumors. However, CAR-T efficacy remains limited due to its exhaustion and shortened persistence. Transcription factors and epigenetic modifications play pivotal roles in modulating T cell differentiation and functionality, which have been leveraged in numerous strategies to promote the formation of long-lasting memory cells with stem-like properties and supercharging CAR-T performance. This review highlights pivotal transcriptional factors, such as c-Jun and FOXO1, which enhance and sustain T cell effector function, diminishes exhaustion, and epigenetic regulators like TET2 and DNMT3A, whose knockout promotes memory T subsets formation. We explore their interconnections, downstream targets, biological impacts, and the potential application risks of certain candidates, providing a comprehensive theoretical framework for supercharging CAR-T therapies through transcriptional and epigenetic interventions.
Collapse
Affiliation(s)
- Diyuan Qin
- Cancer Center, Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A ∗ STAR), Singapore 138673, Singapore
| | - Yanna Lei
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Pei Shu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yugu Zhang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuin-Han Loh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A ∗ STAR), Singapore 138673, Singapore
| | - Yongsheng Wang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Cancer Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qijing Li
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A ∗ STAR), Singapore 138673, Singapore
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
| |
Collapse
|
10
|
Sun Y, Sen S, Parmar R, Arakawa-Hoyt J, Cappelletti M, Rossetti M, Gjertson DW, Sigdel TK, Sarwal MM, Schaenman JM, Bunnapradist S, Lanier LL, Pickering H, Reed EF. Cytotoxic KLRG1+ IL-7R- effector CD8+ T cells distinguish kidney transplant recipients controlling cytomegalovirus reactivation. Front Immunol 2025; 16:1542531. [PMID: 40028342 PMCID: PMC11868092 DOI: 10.3389/fimmu.2025.1542531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction Cytomegalovirus (CMV) viremia remains a major contributor to clinical complications in solid organ transplant (SOT) patients, including organ injury, morbidity and mortality. Given their critical role in antiviral defense, CD8+ T cells are essential for protective immunity against CMV. Methods Using single-cell RNA sequencing, we investigated the transcriptional signatures and developmental lineages of CD8+ T cells in eight immunosuppressed kidney transplant recipients (KTRs) who received organs from CMV-seropositive donors. Results were validated in a cohort of 62 KTRs using immunophenotyping. Results Our data revealed a significant influence of CMV serostatus on transcriptional variance of CD8+ memory T cells, associating with the first principal component from a global analysis of CD8+ T cells (p =0.0406), forming a continuum with five principal differentiation trajectories driven by CMV primary infection or reactivation. Following CMV primary infection, CD8+ T cells were hallmarked by restrained effector-memory differentiation. CD8+ T cells during CMV reactivation diverged non-linearly into senescent-like cells with signatures of arrested cell cycle, diminished translational activity and downregulated ZNF683 and longitudinally expanding effector cells with robust cytotoxic potential and upregulated ZNF683, acting as a reservoir for long-lived effector cells supporting long-term protection. Notably, CD28lo KLRG1hi IL-7R (CD127)lo HLA-DRhi CD8+ T cells present prior to the detection of viremia in CMV-seropositive patients emerged as a key feature distinguishing patients who did or did not undergo CMV reactivation after prophylaxis discontinuation (p =0.0163). Frequencies of these cells were also positively correlated with CMV-stimulated secretion of IFN-γ (p =0.0494), TNF-α (p =0.0358), MIP-1α (p =0.0262), MIP-1β (p =0.0043). Discussion These results provide insights into the transcriptional regulation that influences the generation of CD8+ T cell immunity to CMV and may inform strategics for monitoring host immune response to CMV to better identify and introduce therapeutic intervention to patients at risk of developing clinically significant CMV viremia.
Collapse
Affiliation(s)
- Yumeng Sun
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Subha Sen
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rajesh Parmar
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Janice Arakawa-Hoyt
- Department of Microbiology and Immunology, Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, United States
| | - Monica Cappelletti
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Maura Rossetti
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - David W. Gjertson
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Tara K. Sigdel
- Department of Surgery, Division of Multi Organ Transplantation, University of California, San Francisco, San Francisco, CA, United States
| | - Minnie M. Sarwal
- Department of Surgery, Division of Multi Organ Transplantation, University of California, San Francisco, San Francisco, CA, United States
| | - Joanna M. Schaenman
- Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Suphamai Bunnapradist
- Division of Nephrology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lewis L. Lanier
- Department of Microbiology and Immunology, Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, United States
| | - Harry Pickering
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
11
|
Huo Y, Wang D, Yang S, Xu Y, Qin G, Zhao C, Lei Q, Zhao Q, Liu Y, Guo K, Ouyang S, Sun T, Wang H, Fan F, Han N, Liu H, Chen H, Miao L, Liu L, Duan Y, Lv W, Liu L, Zhang Z, Cang S, Wang L, Zhang Y. Optimal timing of anti-PD-1 antibody combined with chemotherapy administration in patients with NSCLC. J Immunother Cancer 2024; 12:e009627. [PMID: 39706602 PMCID: PMC11667274 DOI: 10.1136/jitc-2024-009627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 11/17/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Anti-programmed cell death 1 (PD-1) antibody combined with chemotherapy simultaneously is regarded as the standard treatment for patients with advanced non-small cell lung cancer (NSCLC) by current clinical guidelines. Different immune statuses induced by chemotherapy considerably affect the synergistic effects of the chemo-anti-PD-1 combination. Therefore, it is necessary to determine the optimal timing of combination treatment administration. METHODS The dynamic immune status induced by chemotherapy was observed in paired peripheral blood samples of patients with NSCLC using flow cytometry and RNA sequencing. Ex vivo studies and metastatic lung carcinoma mouse models were used to evaluate immune activity and explore the optimal combination timing. A multicenter prospective clinical study of 170 patients with advanced NSCLC was performed to assess clinical responses, and systemic immunity was assessed using omics approaches. RESULTS PD-1 expression on CD8+ T cells was downregulated on day 1 (D1) and D2, but recovered on D3 after chemotherapy administration, which is regulated by the calcium influx-P65 signaling pathway. Programmed cell death 1 ligand 1 expression in myeloid-derived suppressor cells was markedly reduced on D3. RNA sequencing analysis showed that T-cell function began to gradually recover on D3 rather than on D1. In addition, ex vivo and in vivo studies have shown that anti-PD-1 treatment on D3 after chemotherapy may enhance the antitumor response and considerably inhibit tumor growth. Finally, in clinical practice, a 3-day-delay sequential combination enhanced the objective response rate (ORR, 68%) and disease control rate (DCR, 98%) compared with the simultaneous combination (ORR=37%; DCR=81%), and prolonged progression-free survival to a greater extent than the simultaneous combination. The new T-cell receptor clones were effectively expanded, and CD8+ T-cell activity was similarly recovered. CONCLUSIONS A 3-day-delay sequential combination might increase antitumor responses and clinical benefits compared with the simultaneous combination.
Collapse
Affiliation(s)
- Yachang Huo
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dan Wang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuangning Yang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yujie Xu
- Department of Oncology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Guohui Qin
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chenhui Zhao
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qingyang Lei
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qitai Zhao
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yaqing Liu
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Kaiyuan Guo
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Songyun Ouyang
- Department of Respiratory and Critical Care Sleep Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ting Sun
- Department of Respiratory and Critical Care Sleep Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongmin Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feifei Fan
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Na Han
- Department of Oncology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hong Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongjie Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lijun Miao
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuqing Duan
- Department of Tumor Immunotherapy, the Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, Hebei, China
| | - Wei Lv
- Department of Tumor Immunotherapy, the Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, Hebei, China
| | - Lihua Liu
- Department of Tumor Immunotherapy, the Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, Hebei, China
| | - Zhixin Zhang
- Department of Technology, Chengdu ExAb Biotechnology Ltd, Chengdu, Sichuan, China
| | - Shundong Cang
- Department of Oncology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Liping Wang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- School of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
12
|
Xiao J, Wang F, Yan H, Wang B, Su B, Lu X, Zhang T. Memory stem CD8 +T cells in HIV/Mtb mono- and co-infection: characteristics, implications, and clinical significance. Front Cell Infect Microbiol 2024; 14:1485825. [PMID: 39720790 PMCID: PMC11666416 DOI: 10.3389/fcimb.2024.1485825] [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: 08/25/2024] [Accepted: 11/13/2024] [Indexed: 12/26/2024] Open
Abstract
Human immunodeficiency Virus (HIV) and Mycobacterium tuberculosis (Mtb) co-infection presents a significant public health challenge worldwide. Comprehensive assessment of the immune response in HIV/Mtb co-infection is complex and challenging. CD8+T cells play a pivotal role in the adaptive immune response to both HIV and Mtb. The differentiation of CD8+T cells follow a hierarchical pattern, with varying degrees of exhaustion throughout the process. Memory stem T cells (TSCM cells) is at the apex of the memory T lymphocyte system, which has recently emerged as a promising target in immunotherapy. In this context, we discuss the alterations of CD8+TSCM cells in HIV/Mtb mono- and co-infection, their implications and clinical significance, and potential for improving immunotherapy.
Collapse
Affiliation(s)
- Jing Xiao
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Fuchun Wang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Hongxia Yan
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Bo Wang
- Department of Respiratory Medicine, Beijing Fengtai Hospital of Integrated Traditional and Western Medicine, Beijing, China
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xiaofan Lu
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Tong Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Zhu Z, Luo Y, Lou G, Yihunie K, Wizzard S, DeVilbiss AW, Muh S, Ma C, Shinde SS, Hoar J, Hu T, Zhang N, Biswal S, DeBerardinis RJ, Wu T, Yao C. The redox sensor KEAP1 facilitates adaptation of T cells to chronic antigen stimulation by preventing hyperactivation. Sci Immunol 2024; 9:eadk2954. [PMID: 39612322 DOI: 10.1126/sciimmunol.adk2954] [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: 08/13/2023] [Revised: 07/10/2024] [Accepted: 11/04/2024] [Indexed: 12/01/2024]
Abstract
During persistent antigen stimulation, exhausted CD8+ T cells are continuously replenished by self-renewing stem-like T cells. However, how CD8+ T cells adapt to chronic stimulation remains unclear. Here, we show that persistent antigen stimulation primes chromatin for regulation by the redox-sensing KEAP1-NRF2 pathway. Loss of KEAP1 in T cells impaired control of chronic viral infection. T cell-intrinsic KEAP1 suppressed NRF2 to promote expansion and persistence of virus-specific CD8+ T cells, drive a stem-like T cell response, down-regulate immune checkpoint molecules, and limit T cell receptor (TCR) hyperactivation and apoptosis. NRF2 epigenetically derepressed BACH2 targets and opposed a stem-like program driven by BACH2. In exhausted T cells induced by tonic GD2 chimeric antigen receptor (CAR) signaling, the effects of KEAP1 deficiency were rescued by inhibiting proximal TCR signaling. Enhancing mitochondrial oxidation improved the expansion and survival of KEAP1-deficient CD8+ GD2 CAR T cells and up-regulated markers associated with stem-like cells. Thus, the KEAP1-NRF2 axis regulates stem-like CD8+ T cells and long-term T cell immunity during chronic antigen exposure.
Collapse
Affiliation(s)
- Ziang Zhu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Immunology PhD Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ying Luo
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Guohua Lou
- Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kiddist Yihunie
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Cancer Biology PhD Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Safuwra Wizzard
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Immunology PhD Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Andrew W DeVilbiss
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sarah Muh
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chaoyu Ma
- Department of Microbiology, Immunology, & Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Sejal S Shinde
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jonathan Hoar
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Taidou Hu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nu Zhang
- Department of Microbiology, Immunology, & Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| | - Shyam Biswal
- Department of Environmental Health and Engineering, Johns Hopkins School of Public Health, Baltimore, MD 21205, USA
- Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern, Dallas, TX 75225, USA
| | - Tuoqi Wu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Cellular Networks in Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chen Yao
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
14
|
Riquelme-Guzmán C, Stout AJ, Kaplan DL, Flack JE. Unlocking the potential of cultivated meat through cell line engineering. iScience 2024; 27:110877. [PMID: 39351194 PMCID: PMC11440241 DOI: 10.1016/j.isci.2024.110877] [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] [Indexed: 10/04/2024] Open
Abstract
Cultivated meat has the potential to revolutionize food production, but its progress is hindered by fundamental shortcomings of mammalian cells with respect to industrial-scale bioprocesses. Here, we discuss the essential role of cell line engineering in overcoming these limitations, highlighting the balance between the benefits of enhanced cellular traits and the associated regulatory and consumer acceptance challenges. We believe that careful selection of cell engineering strategies, including both genetic and non-genetic modifications, can address this trade-off and is essential to advancing the field.
Collapse
Affiliation(s)
- Camilo Riquelme-Guzmán
- Biomedical Engineering Department, Tufts University Center for Cellular Agriculture, Tufts University, Medford, MA, USA
| | - Andrew J Stout
- Biomedical Engineering Department, Tufts University Center for Cellular Agriculture, Tufts University, Medford, MA, USA
- Deco Labs, Inc., Boston, MA, USA
| | - David L Kaplan
- Biomedical Engineering Department, Tufts University Center for Cellular Agriculture, Tufts University, Medford, MA, USA
| | - Joshua E Flack
- Department of Biotechnology, Delft University of Technology, Delft, the Netherlands
| |
Collapse
|
15
|
Srinivasan S, Armitage J, Nilsson J, Waithman J. Transcriptional rewiring in CD8 + T cells: implications for CAR-T cell therapy against solid tumours. Front Immunol 2024; 15:1412731. [PMID: 39399500 PMCID: PMC11466849 DOI: 10.3389/fimmu.2024.1412731] [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: 04/05/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024] Open
Abstract
T cells engineered to express chimeric-antigen receptors (CAR-T cells) can effectively control relapsed and refractory haematological malignancies in the clinic. However, the successes of CAR-T cell therapy have not been recapitulated in solid tumours due to a range of barriers such as immunosuppression, poor infiltration, and tumour heterogeneity. Numerous strategies are being developed to overcome these barriers, which include improving culture conditions and manufacturing protocols, implementing novel CAR designs, and novel approaches to engineering the T cell phenotype. In this review, we describe the various emerging strategies to improve CAR T cell therapy for solid tumours. We specifically focus on new strategies to modulate cell function and fate that have precipitated from the growing knowledge of transcriptional circuits driving T cell differentiation, with the ultimate goal of driving more productive anti-tumour T cell immunity. Evidence shows that enrichment of particular phenotypic subsets of T cells in the initial cell product correlates to improved therapeutic responses and clinical outcomes. Furthermore, T cell exhaustion and poor persistence are major factors limiting therapeutic efficacy. The latest preclinical work shows that targeting specific master regulators and transcription factors can overcome these key barriers, resulting in superior T cell therapeutic products. This can be achieved by targeting key transcriptional circuits promoting memory-like phenotypes or sustaining key effector functions within the hostile tumour microenvironment. Additional discussion points include emerging considerations for the field such as (i) targeting permutations of transcription factors, (ii) transient expression systems, (iii) tissue specificity, and (iv) expanding this strategy beyond CAR-T cell therapy and cancer.
Collapse
Affiliation(s)
- Shamini Srinivasan
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Jesse Armitage
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
- Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Jonas Nilsson
- Melanoma Discovery Lab, Harry Perkins Institute of Medical Research, Centre of Medical Research, The University of Western Australia, Perth, WA, Australia
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Jason Waithman
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
- Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| |
Collapse
|
16
|
Sun Y, Yinwang E, Wang S, Wang Z, Wang F, Xue Y, Zhang W, Zhao S, Mou H, Chen S, Jin L, Li B, Ye Z. Phenotypic and spatial heterogeneity of CD8 + tumour infiltrating lymphocytes. Mol Cancer 2024; 23:193. [PMID: 39251981 PMCID: PMC11382426 DOI: 10.1186/s12943-024-02104-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
CD8+ T cells are the workhorses executing adaptive anti-tumour response, and targets of various cancer immunotherapies. Latest advances have unearthed the sheer heterogeneity of CD8+ tumour infiltrating lymphocytes, and made it increasingly clear that the bulk of the endogenous and therapeutically induced tumour-suppressive momentum hinges on a particular selection of CD8+ T cells with advantageous attributes, namely the memory and stem-like exhausted subsets. A scrutiny of the contemporary perception of CD8+ T cells in cancer and the subgroups of interest along with the factors arbitrating their infiltration contextures, presented herein, may serve as the groundwork for future endeavours to probe further into the regulatory networks underlying their differentiation and migration, and optimise T cell-based immunotherapies accordingly.
Collapse
Affiliation(s)
- Yikan Sun
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Eloy Yinwang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Shengdong Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Zenan Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Fangqian Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Yucheng Xue
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Wenkan Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Shenzhi Zhao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Haochen Mou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Shixin Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Lingxiao Jin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Binghao Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China.
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China.
| | - Zhaoming Ye
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China.
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
17
|
Steiner C, Denlinger N, Huang X, Yang Y. Stem-like CD8 + T cells in cancer. Front Immunol 2024; 15:1426418. [PMID: 39211052 PMCID: PMC11357971 DOI: 10.3389/fimmu.2024.1426418] [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: 05/01/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Stem-like CD8+ T cells (TSL) are a subset of immune cells with superior persistence and antitumor immunity. They are TCF1+ PD-1+ and important for the expansion of tumor specific CD8+ T cells in response to checkpoint blockade immunotherapy. In acute infections, naïve CD8+ T cells differentiate into effector and memory CD8+ T cells; in cancer and chronic infections, persistent antigen stimulation can lead to T cell exhaustion. Recent studies have highlighted the dichotomy between late dysfunctional (or exhausted) T cells (TLD) that are TCF1- PD-1+ and self-renewing TCF1+ PD-1+ TSL from which they derive. TCF1+ TSL cells are considered to have stem cell-like properties akin to memory T cell populations and can give rise to cytotoxic effector and transitory T cell phenotypes (TTE) which mediate tumor control. In this review, we will discuss recent advances made in research on the formation and expansion of TSL, as well as distinct niches required for their differentiation and maintenance in the setting of cancer. We will also discuss potential strategies to generate these cells, with clinical implications for stemness enhancement in vaccine design, immune checkpoint blockade (ICB), and adoptive T cell therapies.
Collapse
Affiliation(s)
| | | | - Xiaopei Huang
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Yiping Yang
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| |
Collapse
|
18
|
Zebley CC, Zehn D, Gottschalk S, Chi H. T cell dysfunction and therapeutic intervention in cancer. Nat Immunol 2024; 25:1344-1354. [PMID: 39025962 PMCID: PMC11616736 DOI: 10.1038/s41590-024-01896-9] [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: 04/12/2024] [Accepted: 06/12/2024] [Indexed: 07/20/2024]
Abstract
Recent advances in immunotherapy have affirmed the curative potential of T cell-based approaches for treating relapsed and refractory cancers. However, the therapeutic efficacy is limited in part owing to the ability of cancers to evade immunosurveillance and adapt to immunological pressure. In this Review, we provide a brief overview of cancer-mediated immunosuppressive mechanisms with a specific focus on the repression of the surveillance and effector function of T cells. We discuss CD8+ T cell exhaustion and functional heterogeneity and describe strategies for targeting the molecular checkpoints that restrict T cell differentiation and effector function to bolster immunotherapeutic effects. We also delineate the emerging contributions of the tumor microenvironment to T cell metabolism and conclude by highlighting discovery-based approaches for developing future cellular therapies. Continued exploration of T cell biology and engineering hold great promise for advancing therapeutic interventions for cancer.
Collapse
Affiliation(s)
- Caitlin C Zebley
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Dietmar Zehn
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan and Center for Infection Prevention (ZIP), Technical University of Munich, Freising, Germany
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
19
|
Dou Z, Bonacci TR, Shou P, Landoni E, Woodcock MG, Sun C, Savoldo B, Herring LE, Emanuele MJ, Song F, Baldwin AS, Wan Y, Dotti G, Zhou X. 4-1BB-encoding CAR causes cell death via sequestration of the ubiquitin-modifying enzyme A20. Cell Mol Immunol 2024; 21:905-917. [PMID: 38937625 PMCID: PMC11291893 DOI: 10.1038/s41423-024-01198-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: 11/03/2023] [Accepted: 06/14/2024] [Indexed: 06/29/2024] Open
Abstract
CD28 and 4-1BB costimulatory endodomains included in chimeric antigen receptor (CAR) molecules play a critical role in promoting sustained antitumor activity of CAR-T cells. However, the molecular events associated with the ectopic and constitutive display of either CD28 or 4-1BB in CAR-T cells have been only partially explored. In the current study, we demonstrated that 4-1BB incorporated within the CAR leads to cell cluster formation and cell death in the forms of both apoptosis and necroptosis in the absence of CAR tonic signaling. Mechanistic studies illustrate that 4-1BB sequesters A20 to the cell membrane in a TRAF-dependent manner causing A20 functional deficiency that in turn leads to NF-κB hyperactivity, cell aggregation via ICAM-1 overexpression, and cell death including necroptosis via RIPK1/RIPK3/MLKL pathway. Genetic modulations obtained by either overexpressing A20 or releasing A20 from 4-1BB by deleting the TRAF-binding motifs of 4-1BB rescue cell cluster formation and cell death and enhance the antitumor ability of 4-1BB-costimulated CAR-T cells.
Collapse
Affiliation(s)
- Zhangqi Dou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | | | - Peishun Shou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Elisa Landoni
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Mark G Woodcock
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Division of Oncology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chuang Sun
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC, USA
| | - Laura E Herring
- Michael Hooker Proteomics Center, Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Michael J Emanuele
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Feifei Song
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Albert S Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Yisong Wan
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.
| | - Xin Zhou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
20
|
Xiong D, Yu H, Sun ZJ. Unlocking T cell exhaustion: Insights and implications for CAR-T cell therapy. Acta Pharm Sin B 2024; 14:3416-3431. [PMID: 39220881 PMCID: PMC11365448 DOI: 10.1016/j.apsb.2024.04.022] [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: 01/28/2024] [Revised: 03/20/2024] [Accepted: 04/01/2024] [Indexed: 09/04/2024] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy as a form of adoptive cell therapy (ACT) has shown significant promise in cancer treatment, demonstrated by the FDA-approved CAR-T cell therapies targeting CD19 or B cell maturation antigen (BCMA) for hematological malignancies, albeit with moderate outcomes in solid tumors. However, despite these advancements, the efficacy of CAR-T therapy is often compromised by T cell exhaustion, a phenomenon that impedes the persistence and effector function of CAR-T cells, leading to a relapse rate of up to 75% in patients treated with CD19 or CD22 CAR-T cells for hematological malignancies. Strategies to overcome CAR-T exhaustion employ state-of-the-art genomic engineering tools and single-cell sequencing technologies. In this review, we provide a comprehensive understanding of the latest mechanistic insights into T cell exhaustion and their implications for the current efforts to optimize CAR-T cell therapy. These insights, combined with lessons learned from benchmarking CAR-T based products in recent clinical trials, aim to address the challenges posed by T cell exhaustion, potentially setting the stage for the development of tailored next-generation approaches to cancer treatment.
Collapse
Affiliation(s)
- Dian Xiong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Science, Wuhan University, Wuhan 430079, China
| | - Haijun Yu
- Department of Radiation and Medical Oncology, Hubei Province Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Science, Wuhan University, Wuhan 430079, China
| |
Collapse
|
21
|
Ali A, Zhang Y, DiPersio J. The invisible hand: How epigenetics shapes CAR T cell destiny. Mol Ther 2024; 32:1614-1616. [PMID: 38761798 PMCID: PMC11184405 DOI: 10.1016/j.ymthe.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/04/2024] [Accepted: 05/06/2024] [Indexed: 05/20/2024] Open
Affiliation(s)
- Alaa Ali
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA.
| | - Yi Zhang
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA; Center for Discovery and Innovation, Nutley, NJ, USA
| | - John DiPersio
- Washington University in Saint Louis, Saint Louis, MO, USA
| |
Collapse
|
22
|
Liao YM, Hsu SH, Chiou SS. Harnessing the Transcriptional Signatures of CAR-T-Cells and Leukemia/Lymphoma Using Single-Cell Sequencing Technologies. Int J Mol Sci 2024; 25:2416. [PMID: 38397092 PMCID: PMC10889174 DOI: 10.3390/ijms25042416] [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: 12/22/2023] [Revised: 02/02/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T-cell therapy has greatly improved outcomes for patients with relapsed or refractory hematological malignancies. However, challenges such as treatment resistance, relapse, and severe toxicity still hinder its widespread clinical application. Traditional transcriptome analysis has provided limited insights into the complex transcriptional landscape of both leukemia cells and engineered CAR-T-cells, as well as their interactions within the tumor microenvironment. However, with the advent of single-cell sequencing techniques, a paradigm shift has occurred, providing robust tools to unravel the complexities of these factors. These techniques enable an unbiased analysis of cellular heterogeneity and molecular patterns. These insights are invaluable for precise receptor design, guiding gene-based T-cell modification, and optimizing manufacturing conditions. Consequently, this review utilizes modern single-cell sequencing techniques to clarify the transcriptional intricacies of leukemia cells and CAR-Ts. The aim of this manuscript is to discuss the potential mechanisms that contribute to the clinical failures of CAR-T immunotherapy. We examine the biological characteristics of CAR-Ts, the mechanisms that govern clinical responses, and the intricacies of adverse events. By exploring these aspects, we hope to gain a deeper understanding of CAR-T therapy, which will ultimately lead to improved clinical outcomes and broader therapeutic applications.
Collapse
Affiliation(s)
- Yu-Mei Liao
- Division of Hematology-Oncology, Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Shih-Hsien Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center of Applied Genomics, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Shyh-Shin Chiou
- Division of Hematology-Oncology, Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center of Applied Genomics, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
23
|
Casucci M, Bonini C, Ruggiero E. Epigenetic checkpoints regulate the fate and function of CAR-T cells. Nat Immunol 2024; 25:4-6. [PMID: 38168961 DOI: 10.1038/s41590-023-01708-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Affiliation(s)
- Monica Casucci
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Bonini
- Università Vita-Salute San Raffaele, Milano, Italy
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Eliana Ruggiero
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|