1
|
Gao Y, He J, Wang J, Xu H, Ma L. Chimeric antigen receptor T cell immunotherapy for gynecological malignancies. Crit Rev Oncol Hematol 2025; 209:104680. [PMID: 40024355 DOI: 10.1016/j.critrevonc.2025.104680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
Gynecologic malignancies pose a serious threat to women's health worldwide. Although immunotherapy has significantly revolutionized cancer treatment strategies, effective therapeutic options for recurrent or advanced gynecologic malignancies are still deficient, posing significant challenges to clinical therapy. Chimeric antigen receptor (CAR) T cell therapy has achieved remarkable efficacy in treating hematologic malignancies, marking a significant change in the oncology treatment paradigm. However, despite the gradual increase in CAR T cell therapy used in treating solid tumors in recent years, its efficacy in treating gynecologic malignancies still needs further validation. This review will thoroughly examine CAR-T cell engineering and its mechanism of action on specific antigens associated with gynecologic malignancies, systematically assess the current application of CAR T cell therapy in gynecologic tumors and the advancements in clinical trials, and discuss the significant challenges and corresponding strategies, thereby offering a scientific foundation and guidance for future research in this area.
Collapse
Affiliation(s)
- Yajuan Gao
- Department of Gynecology and Obstetrics, Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang 310008, China.
| | - Jing He
- Department of Emergency, Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang 310008, China
| | - Jing Wang
- Department of Gynecology and Obstetrics, Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang 310008, China
| | - Haiou Xu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang 310006, China
| | - Lin Ma
- Department of Gynecology and Obstetrics, Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang 310008, China.
| |
Collapse
|
2
|
Jiang R, Yang L, Liu X, Xu Y, Han L, Chen Y, Gao G, Wang M, Su T, Li H, Fang L, Sun N, Du H, Zheng J, Wang G. Genetically engineered macrophages reverse the immunosuppressive tumor microenvironment and improve immunotherapeutic efficacy in TNBC. Mol Ther 2025:S1525-0016(25)00198-4. [PMID: 40119517 DOI: 10.1016/j.ymthe.2025.03.024] [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: 09/20/2024] [Revised: 01/21/2025] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
The main challenges in current immunotherapy for triple-negative breast cancer (TNBC) lie in the immunosuppressive tumor microenvironment (TME). Considering tumor-associated macrophages (TAMs) are the most abundant immune cells in the TME, resetting TAMs is a promising strategy for ameliorating the immunosuppressive TME. Here, we developed genetically engineered macrophages (GEMs) with gene-carrying adenoviruses, to maintain the M1-like phenotype and directly deliver the immune regulators interleukin-12 and CXCL9 into local tumors, thereby reversing the immunosuppressive TME. In tumor-bearing mice, GEMs demonstrated targeted enrichment in tumors and successfully reprogramed TAMs to M1-like macrophages. Moreover, GEMs significantly enhanced the accumulation, proliferation, and activation of CD8+ T cells, mature dendritic cells, and natural killer cells within tumors, while diminishing M2-like macrophages, immunosuppressive myeloid-derived suppressor cells, and regulatory T cells. This treatment efficiently suppressed tumor growth. In addition, combination therapy with GEMs and anti-programmed cell death protein 1 further improved interferon-γ+CD8+ T cell percentages and tumor inhibition efficacy in an orthotopic murine TNBC model. Therefore, this study provides a novel strategy for reversing the immunosuppressive TME and improving immunotherapeutic efficacy through live macrophage-mediated gene delivery.
Collapse
Affiliation(s)
- Ranran Jiang
- Department of Oncology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China; Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Liechi Yang
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Xin Liu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Department of Urology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Yujun Xu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Lulu Han
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Yuxin Chen
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Ge Gao
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Meng Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Tong Su
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Huizhong Li
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Lin Fang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Nan Sun
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Hongwei Du
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Junnian Zheng
- Department of Oncology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
3
|
Ticha P, Northey JJ, Kersten K, Velozo HG, Ironside AJ, Zidek M, Drain A, Lakins JN, Chen YY, Tsai KK, Weaver VM. NCOR2 represses MHC class I molecule expression to drive metastatic progression of breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.642060. [PMID: 40161756 PMCID: PMC11952456 DOI: 10.1101/2025.03.10.642060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Metastatic progression depends upon the ability of disseminated tumor cells to evade immune surveillance. MHC molecule expression facilitates T cell recognition and activation to permit the eradication of metastatic tumor cells. We identified nuclear corepressor 2 (NCOR2) as a key epigenetic regulator of MHC class I molecule expression on breast tumor cells. Patients with triple negative breast cancers (TNBC) that expressed high levels of NCOR2 also exhibited reduced metastasis free survival and decreased MHC class I expression, and the metastatic lesions in patients with TNBC had high nuclear NCOR2 and reduced CD8 T cell levels and activity. Genetically and experimentally reducing NCOR2 expression in tumor cells permitted interferon gamma upregulation of MHC class I, and potentiated CD8 T cell activity and induction of apoptosis to repress metastatic progression of disseminated breast cancer cells. These studies provide evidence to support NCOR2 as a targetable epigenetic regulator of metastasis towards which therapies could be developed to reduce patient mortality.
Collapse
Affiliation(s)
- Pavla Ticha
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA
- Current address: Department of Plastic Surgery, 3rd Faculty of Medicine and University Hospital Kralovske Vinohrady, Charles University in Prague, Srobarova 50, 10034, Praha 10, Czech Republic
| | - Jason J. Northey
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kelly Kersten
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
- Current address: Cancer Metabolism and Microenvironment Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Hugo González Velozo
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
- Laboratory of Tumor Microenvironment and Metastasis, Centro Ciencia & Vida, Santiago, Chile
| | | | - Martin Zidek
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Allison Drain
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jonathan N. Lakins
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yunn-Yi Chen
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kelvin K. Tsai
- Laboratory of Advanced Molecular Therapeutics, Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Valerie M. Weaver
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Radiation Oncology, Department of Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
4
|
Jiang H, Limsuwannarot S, Kulhanek KR, Pal A, Rysavy LW, Su L, Labiad O, Testa S, Ogana H, Waghray D, Tao P, Jude KM, Seet CS, Crooks GM, Moding EJ, Garcia KC, Kalbasi A. IL-9 as a naturally orthogonal cytokine with optimal JAK/STAT signaling for engineered T cell therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633105. [PMID: 39868284 PMCID: PMC11760723 DOI: 10.1101/2025.01.15.633105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Arming T cells with a synthetically orthogonal IL-9 receptor (o9R) permits facile engraftment and potent anti-tumor functions. We considered whether the paucity of natural IL-9R expression could be exploited for T cell immunotherapy given that, in mice, high doses of IL-9 were well-tolerated without discernible immune modulation. Compared to o9R, T cells engineered with IL-9R exhibit superior tissue infiltration, stemness, and anti-tumor activity. These qualities are consistent with a stronger JAK/STAT signal, which in addition to STAT1/3/5, unexpectedly includes STAT4 (canonically associated with IL-12 but not common γ-chain cytokines). IL-9R T cells are exquisitely sensitive to perturbations of proximal signaling, including structure-guided attenuation, amplification, and rebalancing of JAK/STAT signals. Biased IL-9R mutants uncover STAT1 as a rheostat between proliferative stem-like and terminally differentiated effector states. In summary, we identify native IL-9/IL-9R as a natural cytokine-receptor pair with near-orthogonal qualities and an optimal JAK/STAT signaling profile for engineered T cell therapy.
Collapse
Affiliation(s)
- Hua Jiang
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine. Stanford, CA, 94305, USA
- These authors contributed equally to this work
| | - Sam Limsuwannarot
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
- These authors contributed equally to this work
| | - Kayla R. Kulhanek
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, 94143, USA
- Stanford Center for Cancer Cell Therapy
- These authors contributed equally to this work
| | - Aastha Pal
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
- These authors contributed equally to this work
| | - Lea W. Rysavy
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
- These authors contributed equally to this work
| | - Leon Su
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine. Stanford, CA, 94305, USA
| | - Ossama Labiad
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
| | - Stefano Testa
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
| | - Heather Ogana
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
| | - Deepa Waghray
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine. Stanford, CA, 94305, USA
| | - Pingdong Tao
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine. Stanford, CA, 94305, USA
| | - Kevin M. Jude
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine. Stanford, CA, 94305, USA
| | - Christopher S. Seet
- Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gay M. Crooks
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Division of Pediatric Hematology-Oncology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Everett J. Moding
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, 94143, USA
| | - K. Christopher Garcia
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine. Stanford, CA, 94305, USA
- Parker Institute for Cancer Immunotherapy, 1 Letterman Drive, Suite D3500, San Francisco, CA 94129, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Anusha Kalbasi
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
- Stanford Center for Cancer Cell Therapy
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, 94143, USA
- Lead contact
| |
Collapse
|
5
|
Khan SH, Choi Y, Veena M, Lee JK, Shin DS. Advances in CAR T cell therapy: antigen selection, modifications, and current trials for solid tumors. Front Immunol 2025; 15:1489827. [PMID: 39835140 PMCID: PMC11743624 DOI: 10.3389/fimmu.2024.1489827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 12/02/2024] [Indexed: 01/22/2025] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has revolutionized the treatment of hematologic malignancies, achieving remarkable clinical success with FDA-approved therapies targeting CD19 and BCMA. However, the extension of these successes to solid tumors remains limited due to several intrinsic challenges, including antigen heterogeneity and immunosuppressive tumor microenvironments. In this review, we provide a comprehensive overview of recent advances in CAR T cell therapy aimed at overcoming these obstacles. We discuss the importance of antigen identification by emphasizing the identification of tumor-specific and tumor-associated antigens and the development of CAR T therapies targeting these antigens. Furthermore, we highlight key structural innovations, including cytokine-armored CARs, protease-regulated CARs, and CARs engineered with chemokine receptors, to enhance tumor infiltration and activity within the immunosuppressive microenvironment. Additionally, novel manufacturing approaches, such as the Sleeping Beauty transposon system, mRNA-based CAR transfection, and in vivo CAR T cell production, are discussed as scalable solution to improve the accessibility of CAR T cell therapies. Finally, we address critical therapeutic limitations, including cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and suboptimal persistence of CAR T cells. An examination of emerging strategies for countering these limitations reveals that CRISPR-Cas9-mediated genetic modifications and combination therapies utilizing checkpoint inhibitors can improve CAR T cell functionality and durability. By integrating insights from preclinical models, clinical trials, and innovative engineering approaches, this review addresses advances in CAR T cell therapies and their performance in solid tumors.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Neoplasms/therapy
- Neoplasms/immunology
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Antigens, Neoplasm/immunology
- Tumor Microenvironment/immunology
- Animals
- Clinical Trials as Topic
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Safwaan H. Khan
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Yeonjoo Choi
- Division of Hematology/Oncology, Veterans Affairs (VA) Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Mysore Veena
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- Division of Hematology/Oncology, Veterans Affairs (VA) Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - John K. Lee
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Daniel Sanghoon Shin
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- Division of Hematology/Oncology, Veterans Affairs (VA) Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| |
Collapse
|
6
|
Wan R, Fu B, Fu X, Liu Z, Simayi N, Fu Y, Liang H, Li C, Huang W. SMAD4 Regulates the Expression of LCK Affecting Chimeric Antigen Receptor-T Cells Proliferation Through PI3K/Akt Signaling Pathway. J Cell Physiol 2025; 240:e31520. [PMID: 39763264 PMCID: PMC11704455 DOI: 10.1002/jcp.31520] [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: 06/17/2024] [Revised: 12/06/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025]
Abstract
The proliferation of CAR-T cells was hindered and cannot play its killing function well in solid tumors. And yet the regulatory mechanism of CAR-T cell proliferation is not fully understood. Here, we showed that recombinant expression of CD19CAR in T cells significantly increased the basal activation level of CAR-T cells and LCK activation. Both LCK and SMAD4 were essential for CAR-T cells proliferation since over-express LCK or SMAD4 significantly promotes CAR-T cells proliferation, while knock-down LCK or SMAD4 expression inhibited the proliferation of CAR-T cells seriously. More cells go into apoptosis when knock-down LCK or SMAD4 expression, and the cell cycle was arrested in G2/M or S phase, respectively. Over-express LCK or SMAD4 significantly promotes phosphorylation of PI3K and Akt, while it was inhibited when cells were treated with PI3K and Akt inhibitors (LY294002 or MK2206). Further mechanism exploration experiments showed that SMAD4 bound on the promoter region of LCK regulating its expression. Taken together, we reported that the transcription factor SMAD4 regulated the expression of LCK and further involved in the PI3K/Akt signaling pathway to affect the proliferation of CAR-T cells.
Collapse
Affiliation(s)
- Rongxue Wan
- Department of Transfusion Medicine, School of BiotechnologySouthern Medical UniversityGuangzhouGuangdongChina
- Guangzhou Blood Center, Institute of Blood Transfusion and HematologyGuangzhou Medical UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Bowen Fu
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
- The Third Affiliated HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Xiaokang Fu
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Zengping Liu
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Nafeisha Simayi
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Yongshui Fu
- Department of Transfusion Medicine, School of BiotechnologySouthern Medical UniversityGuangzhouGuangdongChina
- Guangzhou Blood Center, Institute of Blood Transfusion and HematologyGuangzhou Medical UniversityGuangzhouGuangdongChina
- Institute of Blood Transfusion and HematologyGuangzhou First People's HospitalGuangzhouGuangdongChina
| | - Huaqin Liang
- Guangzhou Blood Center, Institute of Blood Transfusion and HematologyGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Chengyao Li
- Department of Transfusion Medicine, School of BiotechnologySouthern Medical UniversityGuangzhouGuangdongChina
| | - Wenhua Huang
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
7
|
Feng B, Tang L. A new paradigm for cancer immunotherapy: Orchestrating type 1 and type 2 immunity for curative response. Clin Transl Med 2025; 15:e70154. [PMID: 39739593 DOI: 10.1002/ctm2.70154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025] Open
Affiliation(s)
- Bing Feng
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Materials Science & Engineering, EPFL, Lausanne, Switzerland
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, China
| | - Li Tang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Materials Science & Engineering, EPFL, Lausanne, Switzerland
| |
Collapse
|
8
|
Erickson SM, Manning BM, Kumar A, Patel MR. Engineered Cellular Therapies for the Treatment of Thoracic Cancers. Cancers (Basel) 2024; 17:35. [PMID: 39796666 PMCID: PMC11718842 DOI: 10.3390/cancers17010035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Thoracic malignancies (lung cancers and malignant pleural mesothelioma) are prevalent worldwide and are associated with high morbidity and mortality. Effective treatments are needed for patients with advanced disease. Cell therapies are a promising approach to the treatment of advanced cancers that make use of immune effector cells that have the ability to mediate antitumor immune responses. In this review, we discuss the prospect of chimeric antigen receptor-T (CAR-T) cells, natural killer (NK) cells, T cell receptor-engineered (TCR-T) cells, and tumor-infiltrating lymphocytes (TILs) as treatments for thoracic malignancies. CAR-T cells and TILs have proven successful in several hematologic cancers and advanced melanoma, respectively, but outside of melanoma, results have thus far been unsuccessful in most other solid tumors. NK cells and TCR-T cells are additional cell therapy platforms with their own unique advantages and challenges. Obstacles that must be overcome to develop effective cell therapy for these malignancies include selecting an appropriate target antigen, combating immunosuppressive cells and signaling molecules present in the tumor microenvironment, persistence, and delivering a sufficient quantity of antitumor immune cells to the tumor. Induced pluripotent stem cells (iPSCs) offer great promise as a source for both NK and T cell-based therapies due to their unlimited expansion potential. Here, we review clinical trial data, as well as recent basic scientific advances that offer insight into how we may overcome these obstacles, and provide an overview of ongoing trials testing novel strategies to overcome these obstacles.
Collapse
Affiliation(s)
- Spencer M. Erickson
- Internal Medicine Residency Program, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Benjamin M. Manning
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA (A.K.)
| | - Akhilesh Kumar
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA (A.K.)
| | - Manish R. Patel
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA (A.K.)
| |
Collapse
|
9
|
Geils C, Kathrein K. Augmentation of Solid Tumor Immunotherapy With IL-12. J Gene Med 2024; 26:e70000. [PMID: 39618102 PMCID: PMC11609498 DOI: 10.1002/jgm.70000] [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: 05/31/2024] [Revised: 10/15/2024] [Accepted: 11/01/2024] [Indexed: 12/13/2024] Open
Abstract
Immunotherapy describes a class of therapies in which the immune system is manipulated for therapeutic benefit. These treatments include immune checkpoint inhibitors, adoptive cell therapy, and vaccines. For many hematological malignancies, immunotherapy has emerged as an essential treatment component. However, this success has yet to be replicated for solid tumors, which develop advanced physical and molecular mechanisms for suppressing and evading immune destruction. Nevertheless, cytokine immunotherapy presents a potential remedy to these barriers by delivering a proinflammatory immune signal to the tumor and thereby transforming it from immunologically "cold" to "hot." Interleukin-12 (IL-12), one of the most potent proinflammatory cytokines, was initially investigated for this purpose. However, initial murine and human studies in which IL-12 was administered systemically resulted in dangerous immunotoxicity associated with off-target immune activation. As a result, recent studies have employed advanced cell and molecular engineering approaches to reduce IL-12 toxicity while increasing or maintaining its efficacy such that its effective doses can be tolerated in humans. This review highlights such developments and identifies promising future directions.
Collapse
Affiliation(s)
- Christian Geils
- Department of Biological SciencesUniversity of South CarolinaColumbiaSouth CarolinaUSA
| | - Katie L. Kathrein
- Department of Biological SciencesUniversity of South CarolinaColumbiaSouth CarolinaUSA
| |
Collapse
|
10
|
Olivera I, Etxeberria I, Luri-Rey C, Molero-Glez P, Melero I. Regional and intratumoral adoptive T-cell therapy. IMMUNO-ONCOLOGY TECHNOLOGY 2024; 24:100715. [PMID: 39055165 PMCID: PMC11269935 DOI: 10.1016/j.iotech.2024.100715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Adoptive T-cell therapies (ACTs) including tumor-infiltrating lymphocytes and engineered T cells (transgenic T-cell receptor and chimeric antigen receptor T cells), have made an important impact in the field of cancer treatment over the past years. Most of these therapies are typically administered systemically in approaches that facilitate the elimination of hematologic malignancies. Therapeutical efficacy against solid tumors, however, with the exception of tumor-infiltrating lymphocytes against melanoma, remains limited due to several barriers preventing lymphocyte access to the tumor bed. Building upon the experience of regional administration in other immunotherapies, the regional administration of adoptive cell therapies is being assessed to overcome this challenge, granting a first round of access of the transferred T cells to the tumor niche and thereby ensuring their activation and expansion. Intralesional and intracavitary routes of delivery have been tested with promising antitumor objective responses in preclinical and clinical studies. Additionally, several strategies are being developed to further improve T-cell activity after reinfusing them back to the patient such as combinations with other immunotherapy agents or direct engineering of the transferred T cells, achieving long-term immune memory. Clinical trials testing different regional adoptive T-cell therapies are ongoing but some issues related to methodology of administration and correct selection of the target antigen to avoid on-target/off-tumor side-effects need to be further evaluated and improved. Herein, we discuss the current preclinical and clinical landscape of intratumoral and locoregional delivery of adoptive T-cell therapies.
Collapse
Affiliation(s)
- I. Olivera
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - I. Etxeberria
- Human Oncology and Pathogenesis Program (HOPP), Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York
- Parker Institute for Cancer Immunotherapy, New York, USA
| | - C. Luri-Rey
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - P. Molero-Glez
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - I. Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona
- Department of Oncology, Clínica Universidad de Navarra, Madrid
- Centro del Cancer de la Universidad de Navarra (CCUN), Pamplona, Spain
- Nuffield Department of Medicine (NDM), University of Oxford, Oxford, UK
| |
Collapse
|
11
|
Chang Y, Chang M, Bao X, Dong C. Advancements in adoptive CAR immune cell immunotherapy synergistically combined with multimodal approaches for tumor treatment. Bioact Mater 2024; 42:379-403. [PMID: 39308543 PMCID: PMC11415837 DOI: 10.1016/j.bioactmat.2024.08.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/26/2024] [Accepted: 08/31/2024] [Indexed: 09/25/2024] Open
Abstract
Adoptive immunotherapy, notably involving chimeric antigen receptor (CAR)-T cells, has obtained Food and Drug Administration (FDA) approval as a treatment for various hematological malignancies, demonstrating promising preclinical efficacy against cancers. However, the intricate and resource-intensive autologous cell processing, encompassing collection, expansion, engineering, isolation, and administration, hamper the efficacy of this therapeutic modality. Furthermore, conventional CAR T therapy is presently confined to addressing solid tumors due to impediments posed by physical barriers, the potential for cytokine release syndrome, and cellular exhaustion induced by the immunosuppressive and heterogeneous tumor microenvironment. Consequently, a strategic integration of adoptive immunotherapy with synergistic multimodal treatments, such as chemotherapy, radiotherapy, and vaccine therapy etc., emerges as a pivotal approach to surmount these inherent challenges. This collaborative strategy holds the key to addressing the limitations delineated above, thereby facilitating the realization of more precise personalized therapies characterized by heightened therapeutic efficacy. Such synergistic strategy not only serves to mitigate the constraints associated with adoptive immunotherapy but also fosters enhanced clinical applicability, thereby advancing the frontiers of therapeutic precision and effectiveness.
Collapse
Affiliation(s)
- Yun Chang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China
| | - Mingyang Chang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
| | - Xiaoping Bao
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Purdue University Institute for Cancer Research, West Lafayette, IN, 47907, USA
| | - Cheng Dong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China
| |
Collapse
|
12
|
Carcopino C, Erdogan E, Henrich M, Kobold S. Armoring chimeric antigen receptor (CAR) T cells as micropharmacies for cancer therapy. IMMUNO-ONCOLOGY TECHNOLOGY 2024; 24:100739. [PMID: 39711794 PMCID: PMC11659983 DOI: 10.1016/j.iotech.2024.100739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Chimeric antigen receptor (CAR)-T-cell therapy has emerged as a powerful weapon in the fight against cancer. However, its efficacy is often hindered by challenges such as limited tumor penetration, antigen escape, and immune suppression within the tumor microenvironment. This review explores the potential of armored CAR-T cells, or 'micropharmacies', in overcoming these obstacles and enhancing the therapeutic outcomes of adoptive T-cell (ATC) therapy. We delve into the engineering strategies behind these advanced therapies and the mechanisms through which they improve CAR-T-cell efficacy. Additionally, we discuss the latest advancements and research findings in the field, providing a comprehensive understanding of the role of armored CAR-T cells in cancer treatment. Ultimately, this review highlights the promising future of integrating micropharmacies into ATC therapy, paving the way for more effective and targeted cancer treatments.
Collapse
Affiliation(s)
- C. Carcopino
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
| | - E. Erdogan
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
| | - M. Henrich
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
| | - S. Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Heidelberg, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
13
|
Zhang M, Kong J, Yin F, Shi J, Li J, Qiu Z, Yue B, Wang S, Sun N, Lin Q, Fu L, Wang X, Sun X, Gao Y, Jiang Y, Guo R. Optimizing CAR-T cell Culture: Differential effects of IL-2, IL-12, and IL-21 on CAR-T cells. Cytokine 2024; 184:156758. [PMID: 39299100 DOI: 10.1016/j.cyto.2024.156758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/07/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-T therapy has demonstrated sustained clinical remission in numerous hematologic malignancies and has expanded to encompass solid tumors and autoimmune diseases. While progress is being made in establishing optimal culture conditions for CAR-T cells, the identification of the most effective cytokine for promoting their persistence in vitro remains elusive. METHODS Here, we employed scRNA-seq (single-cell RNA sequencing) analysis to investigate the potential alterations in biological processes within CAR-T cells following exposure to cytokines (IL-2, IL-12, and IL-21) and antigens. Transcriptomic changes in diverse CAR-T groups were compared following various treatments, with a focus on epigenetic modifications, metabolic shifts, cellular senescence, and exhaustion. RESULTS Our study reveals that CAR-T cells treated with antigen, IL-2, and IL-12 exhibit signs of exhaustion and senescence, whereas those treated with IL-21 do not display these characteristics. The activities of glycolysis and epigenetic changes were significantly increased by treatments with antigens, IL-2, and IL-12, while IL-21 treatment maintained the oxidative phosphorylation (OXPHOS) of CAR-T cells. CONCLUSIONS Our findings suggest that IL-21 may play a role in preventing senescence and could be utilized in combination with other strategies, such as IL-2 and IL-12, for CAR-T culture.
Collapse
Affiliation(s)
- Mengmeng Zhang
- Translational Medical Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - JingJing Kong
- Translational Medical Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fanxiang Yin
- Translational Medical Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jianxiang Shi
- Precision Medicine Center, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jin Li
- Translational Medical Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zan Qiu
- Precision Medicine Center, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Baohong Yue
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuya Wang
- Department of Blood Transfusion, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Nannan Sun
- Translational Medical Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Quande Lin
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Liyan Fu
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xiaoqian Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xianlei Sun
- Basic Medical Research Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| | - Yanxia Gao
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Yong Jiang
- Henan Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine and Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Rongqun Guo
- Translational Medical Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
14
|
Liu Y, Xiao L, Yang M, Chen X, Liu H, Wang Q, Guo M, Luo J. CAR-armored-cell therapy in solid tumor treatment. J Transl Med 2024; 22:1076. [PMID: 39609705 PMCID: PMC11603843 DOI: 10.1186/s12967-024-05903-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024] Open
Abstract
Over the past decade, chimeric antigen receptor (CAR)-T cell therapy has emerged as a revolutionary immunotherapeutic approach to combat cancer. This therapy constructs a CAR on the surface of T cells through genetic engineering techniques. The CAR is formed from a combination of antibody-derived or ligand-derived domains and T-cell receptor (TCR) domains. This enables T cells to specifically bind to and activate against tumor cells. However, the efficacy of CAR-T cells in solid tumors remains inconclusive due to several challenges such as poor tumor trafficking, infiltration, and the immunosuppressive tumor microenvironment (TME). In response, CAR natural killer (CAR-NK) and CAR macrophages (CAR-M) have been developed as complementary strategies for solid tumors. CAR-NK cells do not require HLA compatibility, demonstrate reduced toxicity, and are thus seen as potential substitutes for CAR-T cells. Furthermore, CAR-M immunotherapy is also being researched and has shown phagocytic capabilities and tumor-antigen presentation. This study discusses the features, advantages, and limitations of CAR-T, CAR-NK, and CAR-M cells in the treatment of solid tumors and suggests prospective solutions for enhancing the efficacy of CAR host-cell-based immunotherapy.
Collapse
Affiliation(s)
- Yan Liu
- Navy Medical University, Shanghai, 200433, China
| | - Lin Xiao
- Navy Medical University, Shanghai, 200433, China
| | | | - Xuemei Chen
- Linyi People's Hospital, Linyi, Shandong, 276000, China
| | - Hongyue Liu
- Navy Medical University, Shanghai, 200433, China
| | - Quanxing Wang
- Navy Medical University, Shanghai, 200433, China
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, China
| | - Meng Guo
- Navy Medical University, Shanghai, 200433, China.
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, China.
| | - Jianhua Luo
- Navy Medical University, Shanghai, 200433, China.
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, China.
| |
Collapse
|
15
|
Chen F, Wu K, Lin S, Cui J, Chen X, Zeng Z, Yuan N, Fang M, Liu X, Chen Y, Luo W. Enhancing Tumor Immunity with IL-12 and PD-1 Blockade: A Strategy for Inducing Robust Central Memory T Cell Responses in Resistant Cancer Model. Antibodies (Basel) 2024; 13:94. [PMID: 39584994 PMCID: PMC11586976 DOI: 10.3390/antib13040094] [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: 06/11/2024] [Revised: 10/09/2024] [Accepted: 10/18/2024] [Indexed: 11/26/2024] Open
Abstract
Background: Although immune checkpoint inhibitors (ICIs) have demonstrated efficacy in treating advanced cancers, their therapeutic success remains limited for many patients, with initial responders often experiencing resistance and relapse. Interleukin-12 (IL-12) is a powerful cytokine for antitumor immunotherapy, enhancing both lymphocyte recruitment into tumors and immune cell activation. Methods: In this study, we successfully produced mouse interleukin-12 (mIL12) through eukaryotic recombinant expression. In vivo, mIL12 exhibited significant control of tumor immunity in ICI-resistant and aggressive tumor models. Further mechanistic analysis indicated that treatment with mIL12 led to a substantial increase in tumor-infiltrating CD4+ T, CD8+ T, cDC1, and CD103+ cDC1 cells. Results: Our data underscore the potential of a combined therapeutic strategy involving IL-12 with PD-1 and CTLA-4 blockade to elicit a potent antitumor immune response. Notably, the co-administration of mIL12 and PD-1 blockade significantly enhanced the presence of central memory T cells (TCM) within tumors. Conclusions: This study is the first to provide evidence that the combination of mIL12 and PD-1 blockers promotes the generation of TCM, potentially contributing to a robust and durable antitumor effect.
Collapse
Affiliation(s)
- Fentian Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, China; (F.C.); (X.L.)
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, China
| | - Kexin Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, China; (F.C.); (X.L.)
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, China
| | - Shiqi Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, China; (F.C.); (X.L.)
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, China
| | - Jinlong Cui
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, China; (F.C.); (X.L.)
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, China
| | - Xiaoqing Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, China; (F.C.); (X.L.)
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, China
| | - Zhiren Zeng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, China; (F.C.); (X.L.)
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, China
| | - Na Yuan
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, China; (F.C.); (X.L.)
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, China
| | - Mujin Fang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, China; (F.C.); (X.L.)
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, China
| | - Xue Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, China; (F.C.); (X.L.)
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, China
| | - Yuanzhi Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, China; (F.C.); (X.L.)
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, China
| | - Wenxin Luo
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, China; (F.C.); (X.L.)
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, China
| |
Collapse
|
16
|
Ai K, Liu B, Chen X, Huang C, Yang L, Zhang W, Weng J, Du X, Wu K, Lai P. Optimizing CAR-T cell therapy for solid tumors: current challenges and potential strategies. J Hematol Oncol 2024; 17:105. [PMID: 39501358 PMCID: PMC11539560 DOI: 10.1186/s13045-024-01625-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy demonstrates substantial efficacy in various hematological malignancies. However, its application in solid tumors is still limited. Clinical studies report suboptimal outcomes such as reduced cytotoxicity of CAR-T cells and tumor evasion, underscoring the need to address the challenges of sliding cytotoxicity in CAR-T cells. Despite improvements from fourth and next-generation CAR-T cells, new challenges include systemic toxicity from continuously secreted proteins, low productivity, and elevated costs. Recent research targets genetic modifications to boost killing potential, metabolic interventions to hinder tumor progression, and diverse combination strategies to enhance CAR-T cell therapy. Efforts to reduce the duration and cost of CAR-T cell therapy include developing allogenic and in-vivo approaches, promising significant future advancements. Concurrently, innovative technologies and platforms enhance the potential of CAR-T cell therapy to overcome limitations in treating solid tumors. This review explores strategies to optimize CAR-T cell therapies for solid tumors, focusing on enhancing cytotoxicity and overcoming application restrictions. We summarize recent advances in T cell subset selection, CAR-T structural modifications, infiltration enhancement, genetic and metabolic interventions, production optimization, and the integration of novel technologies, presenting therapeutic approaches that could improve CAR-T cell therapy's efficacy and applicability in solid tumors.
Collapse
Affiliation(s)
- Kexin Ai
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Bowen Liu
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China
| | - Xiaomei Chen
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China
| | - Chuxin Huang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Liping Yang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Weiya Zhang
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
| | - Jianyu Weng
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China
| | - Xin Du
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
- Cancer Center, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.
| | - Peilong Lai
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China.
| |
Collapse
|
17
|
Qu R, Zhao Y, Zhang Y. The mechanism of cytokine regulation of cancer occurrence and development in the tumor microenvironment and its application in cancer treatment: a narrative review. Transl Cancer Res 2024; 13:5649-5663. [PMID: 39525000 PMCID: PMC11543031 DOI: 10.21037/tcr-24-679] [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: 08/01/2024] [Accepted: 09/06/2024] [Indexed: 11/16/2024]
Abstract
Background and Objective The occurrence and development of tumors in human tissues widely depend on their surrounding environment, known as the tumor microenvironment (TME), which comprises various cells, molecules, and blood vessels. Through modifications, organization, and integration, these elements serve as potential therapeutic targets in anti-cancer therapy, supporting and promoting the proliferation, invasion, and metabolism of tumor cells. Cytokines within TME are responsible for immune cell activation, proliferation, and differentiation, thereby influencing the tumor's behavior. This article reviews the use of cytokines in tumor immunotherapy and combs the network signals that cytokines mediate in the development of malignancies. Methods A literature search of international sources was carried out on the PubMed and Web of Science databases, using main keywords such as "tumor immunotherapy", "cytokines", "chemokines", "tumor microenvironment", "recombinant cytokine engineering", and "tumor necrosis factor superfamily". Key Content and Findings The review provides a thorough summary of the functions of tumor necrosis factor superfamilies, chemokines, and interleukins within the TME as well as their therapeutic uses. Their potential as novel targets for tumor treatment is also evaluated. Furthermore, this paper focuses on various feasible strategies for recombinant cytokines reported in recent years, especially the cytokine engineering methods for targeting tumors. Ultimately, this paper contributes to an enhanced understanding among researchers of the mechanisms underlying the impact of the TME on disease development, thereby laying a solid foundation for the future development of new tumor therapies based on cytokines within the TME. Conclusions Cytokine immunotherapy holds promise on antitumor therapy. It is anticipated that the effectiveness of tumor treatment and the quality of life for tumor patients will continue to improve with ongoing research and development in this field.
Collapse
Affiliation(s)
- Run Qu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dali University, Dali, China
| | - Yanhong Zhao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dali University, Dali, China
| | - Yuzhe Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dali University, Dali, China
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, China
| |
Collapse
|
18
|
Zhou A, Jia J, Ji X, Cheng S, Song X, Hu J, Zhao Y, Yu L, Wang J, Wang F. Reshaped Local and Systemic Immune Responses Triggered by a Biomimetic Multifunctional Nanoplatform Coordinating Multi-Pathways for Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39356986 DOI: 10.1021/acsami.4c05714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Immunotherapy has fundamentally transformed the clinical cancer treatment landscape; however, achieving intricate and multifaceted modulation of the immune systems remains challenging. Here, a multipathway coordination of immunogenic cell death (ICD), autophagy, and indoleamine 2,3-dioxygenase-1 (IDO1) was achieved by a biomimetic nano-immunomodulator assembled from a chemotherapeutic agent (doxorubicin, DOX), small interfering RNA (siRNA) molecules targeting IDO1 (siIDO1), and the zeolitic imidazolate framework-8 (ZIF-8). After being camouflaged with a macrophage membrane, the biomimetic nanosystem, named mRDZ, enriched in tumors, which allowed synergistic actions of its components within tumor cells. The chemotherapeutic intervention led to a compensatory upregulation in the expression of IDO1, consequently exerting an inhibitory effect on the reactive oxygen species (ROS) and autophagic responses triggered by DOX and ZIF-8. Precise gene silencing of IDO1 by siIDO1 alleviated its suppressive influence, thereby facilitating increased ROS production and improved autophagy, ultimately bolstering tumor immunogenicity. mRDZ exhibited strong capability to boost potent local and systemic antitumor immune responses with a feature of memory, which led to the effective suppression of the growth, lung metastasis, and recurrence of the tumor. Serving as an exemplary model for the straightforward and potent reshaping of the immune system against tumors, mRDZ offers valuable insights into the development of immunomodulatory nanomaterials for cancer therapy.
Collapse
Affiliation(s)
- Ao Zhou
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Jingyan Jia
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Xueyang Ji
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Sunying Cheng
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xiaoxin Song
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jingyan Hu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yan Zhao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Luying Yu
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Jieting Wang
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Fang Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
19
|
Feng B, Bai Z, Zhou X, Zhao Y, Xie YQ, Huang X, Liu Y, Enbar T, Li R, Wang Y, Gao M, Bonati L, Peng MW, Li W, Tao B, Charmoy M, Held W, Melenhorst JJ, Fan R, Guo Y, Tang L. The type 2 cytokine Fc-IL-4 revitalizes exhausted CD8 + T cells against cancer. Nature 2024; 634:712-720. [PMID: 39322665 PMCID: PMC11485240 DOI: 10.1038/s41586-024-07962-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/20/2024] [Indexed: 09/27/2024]
Abstract
Current cancer immunotherapy predominately focuses on eliciting type 1 immune responses fighting cancer; however, long-term complete remission remains uncommon1,2. A pivotal question arises as to whether type 2 immunity can be orchestrated alongside type 1-centric immunotherapy to achieve enduring response against cancer3,4. Here we show that an interleukin-4 fusion protein (Fc-IL-4), a typical type 2 cytokine, directly acts on CD8+ T cells and enriches functional terminally exhausted CD8+ T (CD8+ TTE) cells in the tumour. Consequently, Fc-IL-4 enhances antitumour efficacy of type 1 immunity-centric adoptive T cell transfer or immune checkpoint blockade therapies and induces durable remission across several syngeneic and xenograft tumour models. Mechanistically, we discovered that Fc-IL-4 signals through both signal transducer and activator of transcription 6 (STAT6) and mammalian target of rapamycin (mTOR) pathways, augmenting the glycolytic metabolism and the nicotinamide adenine dinucleotide (NAD) concentration of CD8+ TTE cells in a lactate dehydrogenase A-dependent manner. The metabolic modulation mediated by Fc-IL-4 is indispensable for reinvigorating intratumoural CD8+ TTE cells. These findings underscore Fc-IL-4 as a potent type 2 cytokine-based immunotherapy that synergizes effectively with type 1 immunity to elicit long-lasting responses against cancer. Our study not only sheds light on the synergy between these two types of immune responses, but also unveils an innovative strategy for advancing next-generation cancer immunotherapy by integrating type 2 immune factors.
Collapse
Affiliation(s)
- Bing Feng
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Materials Science and Engineering, EPFL, Lausanne, Switzerland
| | - Zhiliang Bai
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Xiaolei Zhou
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Materials Science and Engineering, EPFL, Lausanne, Switzerland
| | - Yang Zhao
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Yu-Qing Xie
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Xinyi Huang
- Institute of Chemical Sciences and Engineering, EPFL, Lausanne, Switzerland
| | - Yang Liu
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Tom Enbar
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Rongrong Li
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Yi Wang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Materials Science and Engineering, EPFL, Lausanne, Switzerland
| | - Min Gao
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Lucia Bonati
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Mei-Wen Peng
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Weilin Li
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Bo Tao
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Mélanie Charmoy
- Department of Oncology, University of Lausanne, Epalinges, Switzerland
| | - Werner Held
- Department of Oncology, University of Lausanne, Epalinges, Switzerland
| | | | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
| | - Yugang Guo
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Institute of Materials Science and Engineering, EPFL, Lausanne, Switzerland.
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Li Tang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Institute of Materials Science and Engineering, EPFL, Lausanne, Switzerland.
| |
Collapse
|
20
|
Peng L, Sferruzza G, Yang L, Zhou L, Chen S. CAR-T and CAR-NK as cellular cancer immunotherapy for solid tumors. Cell Mol Immunol 2024; 21:1089-1108. [PMID: 39134804 PMCID: PMC11442786 DOI: 10.1038/s41423-024-01207-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/22/2024] [Indexed: 10/02/2024] Open
Abstract
In the past decade, chimeric antigen receptor (CAR)-T cell therapy has emerged as a promising immunotherapeutic approach for combating cancers, demonstrating remarkable efficacy in relapsed/refractory hematological malignancies in both pediatric and adult patients. CAR-natural killer (CAR-NK) cell complements CAR-T cell therapy by offering several distinct advantages. CAR-NK cells do not require HLA compatibility and exhibit low safety concerns. Moreover, CAR-NK cells are conducive to "off-the-shelf" therapeutics, providing significant logistic advantages over CAR-T cells. Both CAR-T and CAR-NK cells have shown consistent and promising results in hematological malignancies. However, their efficacy against solid tumors remains limited due to various obstacles including limited tumor trafficking and infiltration, as well as an immuno-suppressive tumor microenvironment. In this review, we discuss the recent advances and current challenges of CAR-T and CAR-NK cell immunotherapies, with a specific focus on the obstacles to their application in solid tumors. We also analyze in depth the advantages and drawbacks of CAR-NK cells compared to CAR-T cells and highlight CAR-NK CAR optimization. Finally, we explore future perspectives of these adoptive immunotherapies, highlighting the increasing contribution of cutting-edge biotechnological tools in shaping the next generation of cellular immunotherapy.
Collapse
Affiliation(s)
- Lei Peng
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
- System Biology Institute, Yale University, West Haven, CT, USA.
| | - Giacomo Sferruzza
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
| | - Luojia Yang
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, CT, USA
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, CT, USA
| | - Liqun Zhou
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, CT, USA
- Immunobiology Program, Yale University, New Haven, CT, USA
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
- System Biology Institute, Yale University, West Haven, CT, USA.
- Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, CT, USA.
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, CT, USA.
- Immunobiology Program, Yale University, New Haven, CT, USA.
- Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA.
- Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA.
- Yale Liver Center, Yale University School of Medicine, New Haven, CT, USA.
- Yale Center for Biomedical Data Science, Yale University School of Medicine, New Haven, CT, USA.
- Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
21
|
Xu EJK, Smith BE, Conce Alberto WD, Walsh MJ, Lim B, Hoffman MT, Qiang L, Dong J, Garmilla A, Zhao QH, Perez CR, Gaglione SA, Dobson CS, Dougan M, Dougan SK, Birnbaum ME. Peptide-MHC-targeted retroviruses enable in vivo expansion and gene delivery to tumor-specific T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.613594. [PMID: 39345591 PMCID: PMC11429759 DOI: 10.1101/2024.09.18.613594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Tumor-infiltrating-lymphocyte (TIL) therapy has demonstrated that endogenous T cells can be harnessed to initiate an effective anti-tumor response. Despite clinical promise, current TIL production protocols involve weeks-long ex vivo expansions which can affect treatment efficacy. Therefore, additional tools are needed to engineer endogenous tumor-specific T cells to have increased potency while mitigating challenges of manufacturing. Here, we present a strategy for pseudotyping retroviral vectors with peptide-major histocompatibility complexes (pMHC) for antigen-specific gene delivery to CD8 T cells and examine the efficacy of these transduced cells in immunocompetent mouse models. We demonstrate that pMHC-targeted viruses are able to specifically deliver function-enhancing cargoes while simultaneously activating and expanding anti-tumor T cells. The specificity of these viral vectors enables in vivo engineering of tumor-specific T cells, circumventing ex vivo manufacturing processes and improving overall survival in B16F10-bearing mice. Altogether, we have established that pMHC-targeted viruses are efficient vectors for reprogramming and expanding tumor-specific populations of T cells directly in vivo , with the potential to substantially streamline engineered cell therapy production for a variety of applications.
Collapse
|
22
|
Erler P, Kurcon T, Cho H, Skinner J, Dixon C, Grudman S, Rozlan S, Dessez E, Mumford B, Jo S, Boyne A, Juillerat A, Duchateau P, Poirot L, Aranda-Orgilles B. Multi-armored allogeneic MUC1 CAR T cells enhance efficacy and safety in triple-negative breast cancer. SCIENCE ADVANCES 2024; 10:eadn9857. [PMID: 39213364 PMCID: PMC11364110 DOI: 10.1126/sciadv.adn9857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
Solid tumors, such as triple-negative breast cancer (TNBC), are biologically complex due to cellular heterogeneity, lack of tumor-specific antigens, and an immunosuppressive tumor microenvironment (TME). These challenges restrain chimeric antigen receptor (CAR) T cell efficacy, underlining the importance of armoring. In solid cancers, a localized tumor mass allows alternative administration routes, such as intratumoral delivery with the potential to improve efficacy and safety but may compromise metastatic-site treatment. Using a multi-layered CAR T cell engineering strategy that allowed a synergy between attributes, we show enhanced cytotoxic activity of MUC1 CAR T cells armored with PD1KO, tumor-specific interleukin-12 release, and TGFBR2KO attributes catered towards the TNBC TME. Intratumoral treatment effectively reduced distant tumors, suggesting retention of antigen-recognition benefits at metastatic sites. Overall, we provide preclinical evidence of armored non-alloreactive MUC1 CAR T cells greatly reducing high TNBC tumor burden in a TGFB1- and PD-L1-rich TME both at local and distant sites while preserving safety.
Collapse
Affiliation(s)
| | | | - Hana Cho
- Cellectis Inc., New York, NY, USA
| | | | | | | | | | | | | | - Sumin Jo
- Cellectis Inc., New York, NY, USA
| | | | | | | | | | | |
Collapse
|
23
|
Ma L, Zhang K, Xu J, Wang J, Jiang T, Du X, Zhang J, Huang J, Ren F, Liu D, Xue W, Kan D, Yao M, Liang Y, Jason-Sun H. Building a novel TRUCK by harnessing the endogenous IFN-gamma promoter for cytokine expression. Mol Ther 2024; 32:2728-2740. [PMID: 38879754 PMCID: PMC11405158 DOI: 10.1016/j.ymthe.2024.06.017] [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: 12/25/2023] [Revised: 04/22/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
Despite the remarkable success of chimeric antigen receptor (CAR) T therapy in hematological malignancies, its efficacy in solid tumors remains limited. Cytokine-engineered CAR T cells offer a promising avenue, yet their clinical translation is hindered by the risks associated with constitutive cytokine expression. In this proof-of-concept study, we leverage the endogenous interferon (IFN)-γ promoter for transgenic interleukin (IL)-15 expression. We demonstrate that IFN-γ expression is tightly regulated by T cell receptor signaling. By introducing an internal ribosome entry site IL15 into the 3' UTR of the IFN-γ gene via homology directed repair-mediated knock-in, we confirm that IL-15 expression can co-express with IFN-γ in an antigen stimulation-dependent manner. Importantly, the insertion of transgenes does not compromise endogenous IFN-γ expression. In vitro and in vivo data demonstrate that IL-15 driven by the IFN-γ promoter dramatically improves CAR T cells' antitumor activity, suggesting the effectiveness of IL-15 expression. Last, as a part of our efforts toward clinical translation, we have developed an innovative two-gene knock-in approach. This approach enables the simultaneous integration of CAR and IL-15 genes into TRAC and IFN-γ gene loci using a single AAV vector. CAR T cells engineered to express IL-15 using this approach demonstrate enhanced antitumor efficacy. Overall, our study underscores the feasibility of utilizing endogenous promoters for transgenic cytokines expression in CAR T cells.
Collapse
MESH Headings
- Interferon-gamma/metabolism
- Promoter Regions, Genetic
- Humans
- Animals
- Mice
- Immunotherapy, Adoptive/methods
- Interleukin-15/genetics
- Interleukin-15/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/immunology
- Genetic Vectors/genetics
- Cell Line, Tumor
- Transgenes
- Cytokines/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Xenograft Model Antitumor Assays
- Gene Expression
Collapse
Affiliation(s)
- Liya Ma
- Shenzhen Celconta Life Science Co. Ltd., Shenzhen, Guangdong, China
| | - Kaiwen Zhang
- Shenzhen Celconta Life Science Co. Ltd., Shenzhen, Guangdong, China
| | - Jian Xu
- Shenzhen Celconta Life Science Co. Ltd., Shenzhen, Guangdong, China
| | - Jian Wang
- Shenzhen Celconta Life Science Co. Ltd., Shenzhen, Guangdong, China
| | - Ting Jiang
- Shenzhen Celconta Life Science Co. Ltd., Shenzhen, Guangdong, China
| | - Xiaolong Du
- Shenzhen Celconta Life Science Co. Ltd., Shenzhen, Guangdong, China
| | - Jiaxin Zhang
- Shenzhen Celconta Life Science Co. Ltd., Shenzhen, Guangdong, China
| | - Jing Huang
- Shenzhen Celconta Life Science Co. Ltd., Shenzhen, Guangdong, China
| | - Fengyi Ren
- Shenzhen Celconta Life Science Co. Ltd., Shenzhen, Guangdong, China
| | - Dong Liu
- Shenzhen Celconta Life Science Co. Ltd., Shenzhen, Guangdong, China
| | - Weiwei Xue
- Shenzhen Celconta Life Science Co. Ltd., Shenzhen, Guangdong, China
| | - Dongxu Kan
- Shenzhen Celconta Life Science Co. Ltd., Shenzhen, Guangdong, China
| | - Mengjiao Yao
- Shenzhen Celconta Life Science Co. Ltd., Shenzhen, Guangdong, China
| | - Yutian Liang
- Shenzhen Celconta Life Science Co. Ltd., Shenzhen, Guangdong, China
| | | |
Collapse
|
24
|
Xin Z, Qu S, Qu Y, Xu Y, Liu R, Sun D, Dai Z. Emerging IL-12-based nanomedicine for cancer therapy. NANO TODAY 2024; 57:102331. [DOI: 10.1016/j.nantod.2024.102331] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
25
|
Xin Q, Chen Y, Sun X, Li R, Wu Y, Huang X. CAR-T therapy for ovarian cancer: Recent advances and future directions. Biochem Pharmacol 2024; 226:116349. [PMID: 38852648 DOI: 10.1016/j.bcp.2024.116349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/21/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
Ovarian cancer (OC) is a common gynecological tumor with high mortality, which is difficult to control its progression with conventional treatments and is prone to recurrence. Recent studies have identified OC as an immunogenic tumor that can be recognized by the host immune system. Immunotherapy for OC is being evaluated, but approaches such as immune checkpoint inhibitors have limited efficacy, adoptive cell therapy is an alternative therapy, in which CAR(chimeric antigen receptor)-T therapy has been applied to the clinical treatment of hematological malignancies. In addition, CAR-NK and CAR-macrophage (CAR-M) have also shown great potential in the treatment of solid tumors. Here, we discuss recent advances in preclinical and clinical studies of CAR-T for OC treatment, introduce the efforts made by researchers to modify the structure of CAR in order to achieve effective OC immunotherapy, as well as the research status of CAR-NK and CAR-M, and highlight emerging therapeutic opportunities that can be utilized to improve the survival of patients with OC using CAR-based adoptive cell therapy.
Collapse
Affiliation(s)
- Qianling Xin
- Anhui Women and Children's Medical Center, Hefei Maternal and Child Health Hospital, Hefei, China
| | - Yizhao Chen
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Xiaojing Sun
- Anhui Women and Children's Medical Center, Hefei Maternal and Child Health Hospital, Hefei, China
| | - Ruilin Li
- Department of Pharmacy, The Third Affiliated Hospital of Anhui Medical University, Hefei First People's Hospital, Hefei, China.
| | - Yujing Wu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China.
| | - Xuegui Huang
- Anhui Women and Children's Medical Center, Hefei Maternal and Child Health Hospital, Hefei, China.
| |
Collapse
|
26
|
Posey AD, Young RM, June CH. Future perspectives on engineered T cells for cancer. Trends Cancer 2024; 10:687-695. [PMID: 38853073 DOI: 10.1016/j.trecan.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 06/11/2024]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has emerged as a revolutionary treatment for hematological malignancies, but its adaptation to solid tumors is impeded by multiple challenges, particularly T cell dysfunction and exhaustion. The heterogeneity and inhospitableness of the solid tumor microenvironment (TME) contribute to diminished CAR T cell efficacy exhibited by reduced cytotoxicity, proliferation, cytokine secretion, and the upregulation of inhibitory receptors, similar to the phenotype of tumor-infiltrating lymphocytes (TILs). In this review, we highlight recent advances in T cell therapy for solid tumors, particularly brain cancer. Innovative strategies, including locoregional delivery and 'armoring' CAR T cells with cytokines such as interleukin (IL)-18, are under investigation to improve efficacy and safety. We also highlight emerging issues with toxicity management of CAR T cell adverse events. This review discusses the obstacles associated with CAR T cell therapy in the context of solid tumors and outlines current and future strategies to overcome these challenges.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Neoplasms/immunology
- Neoplasms/therapy
- Neoplasms/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Microenvironment/immunology
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Animals
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
Collapse
Affiliation(s)
- Avery D Posey
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Parker Institute for Cancer Immunotherapy at the University of Pennsylvania, Philadelphia, PA, USA; Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Regina M Young
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Parker Institute for Cancer Immunotherapy at the University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, 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; Parker Institute for Cancer Immunotherapy at the University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
27
|
Farhangnia P, Khorramdelazad H, Nickho H, Delbandi AA. Current and future immunotherapeutic approaches in pancreatic cancer treatment. J Hematol Oncol 2024; 17:40. [PMID: 38835055 PMCID: PMC11151541 DOI: 10.1186/s13045-024-01561-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024] Open
Abstract
Pancreatic cancer is a major cause of cancer-related death, but despondently, the outlook and prognosis for this resistant type of tumor have remained grim for a long time. Currently, it is extremely challenging to prevent or detect it early enough for effective treatment because patients rarely exhibit symptoms and there are no reliable indicators for detection. Most patients have advanced or spreading cancer that is difficult to treat, and treatments like chemotherapy and radiotherapy can only slightly prolong their life by a few months. Immunotherapy has revolutionized the treatment of pancreatic cancer, yet its effectiveness is limited by the tumor's immunosuppressive and hard-to-reach microenvironment. First, this article explains the immunosuppressive microenvironment of pancreatic cancer and highlights a wide range of immunotherapy options, including therapies involving oncolytic viruses, modified T cells (T-cell receptor [TCR]-engineered and chimeric antigen receptor [CAR] T-cell therapy), CAR natural killer cell therapy, cytokine-induced killer cells, immune checkpoint inhibitors, immunomodulators, cancer vaccines, and strategies targeting myeloid cells in the context of contemporary knowledge and future trends. Lastly, it discusses the main challenges ahead of pancreatic cancer immunotherapy.
Collapse
Affiliation(s)
- Pooya Farhangnia
- Reproductive Sciences and Technology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hamid Nickho
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali-Akbar Delbandi
- Reproductive Sciences and Technology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Goto A, Moriya Y, Nakayama M, Iwasaki S, Yamamoto S. DMPK perspective on quantitative model analysis for chimeric antigen receptor cell therapy: Advances and challenges. Drug Metab Pharmacokinet 2024; 56:101003. [PMID: 38843652 DOI: 10.1016/j.dmpk.2024.101003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/26/2024] [Accepted: 02/10/2024] [Indexed: 06/24/2024]
Abstract
Chimeric antigen receptor (CAR) cells are genetically engineered immune cells that specifically target tumor-associated antigens and have revolutionized cancer treatment, particularly in hematological malignancies, with ongoing investigations into their potential applications in solid tumors. This review provides a comprehensive overview of the current status and challenges in drug metabolism and pharmacokinetics (DMPK) for CAR cell therapy, specifically emphasizing on quantitative modeling and simulation (M&S). Furthermore, the recent advances in quantitative model analysis have been reviewed, ranging from clinical data characterization to mechanism-based modeling that connects in vitro and in vivo nonclinical and clinical study data. Additionally, the future perspectives and areas for improvement in CAR cell therapy translation have been reviewed. This includes using formulation quality considerations, characterization of appropriate animal models, refinement of in vitro models for bottom-up approaches, and enhancement of quantitative bioanalytical methodology. Addressing these challenges within a DMPK framework is pivotal in facilitating the translation of CAR cell therapy, ultimately enhancing the patients' lives through efficient CAR cell therapies.
Collapse
Affiliation(s)
- Akihiko Goto
- Center of Excellence for Drug Metabolism, Pharmacokinetics and Modeling, Preclinical and Translational Sciences, Research, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | - Yuu Moriya
- Center of Excellence for Drug Metabolism, Pharmacokinetics and Modeling, Preclinical and Translational Sciences, Research, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | - Miyu Nakayama
- Center of Excellence for Drug Metabolism, Pharmacokinetics and Modeling, Preclinical and Translational Sciences, Research, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | - Shinji Iwasaki
- Center of Excellence for Drug Metabolism, Pharmacokinetics and Modeling, Preclinical and Translational Sciences, Research, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | - Syunsuke Yamamoto
- Center of Excellence for Drug Metabolism, Pharmacokinetics and Modeling, Preclinical and Translational Sciences, Research, Takeda Pharmaceutical Company Limited, Kanagawa, Japan.
| |
Collapse
|
29
|
Shin SC, Vickman RE, Filimon B, Yang Y, Hu Z, Mangold KA, Prabhakar BS, Schreiber H, Xu W. The safety and efficacy of systemic delivery of a new liver-de-targeted TGFβ signaling inhibiting adenovirus in an immunocompetent triple negative mouse mammary tumor model. Cancer Gene Ther 2024; 31:574-585. [PMID: 38267626 PMCID: PMC11016465 DOI: 10.1038/s41417-024-00735-1] [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: 09/01/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
Aberrant TGFβ signaling is linked to metastasis and tumor immune escape of many cancers including metastatic triple negative breast cancer (mTNBC). Previously, we have found that oncolytic adenoviruses expressing a TGFβ signaling inhibitory protein (sTGFβRIIFc) induced immune activation in a mouse TNBC (4T1) immunocompetent subcutaneous model with intratumoral injection. Systemic administration of adenoviruses can be a superior route to treat mTNBC but faces the challenges of increased toxicity and viral clearance. Thus, we created a liver-de-targeted sTGFβRIIFc- and LyP-1 peptide-expressing adenovirus (mHAdLyp.sT) with enhanced breast cancer cell tropism. Its safety and immune response features were profiled in the 4T1 model. Our data showed that the systemic administration of mHAdLyp.sT resulted in reduced hepatic and systemic toxicity. mHAdLyp.sT was also effective in increasing Th1 cytokines and anti-tumor cell populations by cytokine analysis, spleen/tumor qRT-PCR, and flow cytometry. We further tested the therapeutic effects of mHAdLyp.sT alone and in combination with immune checkpoint inhibitors (ICIs). mHAdLyp.sT alone and with all ICI combinations elicited significant inhibition of lung metastasis by histological analysis. When mHAdLyp.sT was combined with both anti-PD-1 and anti-CTLA-4 antibodies, primary 4T1 tumor growth was also significantly inhibited. We are confident in advancing this new treatment option for mTNBC.
Collapse
Affiliation(s)
- Soon Cheon Shin
- Cancer Gene Therapy Program, Department of Medicine, NorthShore University HealthSystem, an Academic Affiliate of the University of Chicago Pritzker School of Medicine, Endeavor Health Medical Group, Evanston, IL, USA
| | - Renee E Vickman
- Center for Personalized Cancer Care, Department of Surgery, NorthShore University HealthSystem, an Academic Affiliate of the University of Chicago Pritzker School of Medicine, Endeavor Health Medical Group, Evanston, IL, USA
| | - Beniamin Filimon
- Cancer Gene Therapy Program, Department of Medicine, NorthShore University HealthSystem, an Academic Affiliate of the University of Chicago Pritzker School of Medicine, Endeavor Health Medical Group, Evanston, IL, USA
| | - Yuefeng Yang
- Cancer Gene Therapy Program, Department of Medicine, NorthShore University HealthSystem, an Academic Affiliate of the University of Chicago Pritzker School of Medicine, Endeavor Health Medical Group, Evanston, IL, USA
- Department of Experimental Medical Science and Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, China
| | - Zebin Hu
- Cancer Gene Therapy Program, Department of Medicine, NorthShore University HealthSystem, an Academic Affiliate of the University of Chicago Pritzker School of Medicine, Endeavor Health Medical Group, Evanston, IL, USA
- National Institutes for Food and Drug Control, Beijing, China
| | - Kathy A Mangold
- Department of Pathology and Laboratory Medicine, NorthShore University HealthSystem, Endeavor Health Medical Group, Evanston, IL, USA
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Hans Schreiber
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Weidong Xu
- Cancer Gene Therapy Program, Department of Medicine, NorthShore University HealthSystem, an Academic Affiliate of the University of Chicago Pritzker School of Medicine, Endeavor Health Medical Group, Evanston, IL, USA.
| |
Collapse
|
30
|
Minnar CM, Lui G, Gulley JL, Schlom J, Gameiro SR. Preclinical and clinical studies of a tumor targeting IL-12 immunocytokine. Front Oncol 2024; 13:1321318. [PMID: 38260854 PMCID: PMC10802843 DOI: 10.3389/fonc.2023.1321318] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/05/2023] [Indexed: 01/24/2024] Open
Abstract
The clinical success of immune checkpoint inhibitors (ICIs) has demonstrated the promise and challenges of cancer immunotherapy. There is an unmet need to develop novel cancer therapies that can provide clinical benefit for most patients with solid malignancies, which harbor innate or acquired resistance to ICIs. Interleukin-12 (IL-12) is a promising cytokine for cancer therapy given its direct stimulatory effects on innate and adaptive immunity. However, unfavorable pharmacokinetics and a narrow therapeutic index render recombinant IL-12 (rIL-12) less attractive as a cancer therapy. NHS-IL12 is a fusion protein of IL-12 and NHS76 (human IgG1) antibody engineered to target single and double stranded DNA present in necrotic areas solid tumors. In preclinical tumor models, NHS-IL12 elicited significant Th1 immune activation and tumor suppressive effects, primarily mediated by NK and CD8+ T lymphocytes, with engagement of myeloid immunity. NHS-IL12 is currently being evaluated clinically in combination with various therapeutic modalities, including chemotherapy, radiation therapy, immune checkpoint inhibition, vaccines, and epigenetic modulation. Here we review the preclinical and clinical studies involving NHS-IL12 for the treatment of solid malignancies.
Collapse
|
31
|
Luo M, Gong W, Zhang Y, Li H, Ma D, Wu K, Gao Q, Fang Y. New insights into the stemness of adoptively transferred T cells by γc family cytokines. Cell Commun Signal 2023; 21:347. [PMID: 38049832 PMCID: PMC10694921 DOI: 10.1186/s12964-023-01354-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/11/2023] [Indexed: 12/06/2023] Open
Abstract
T cell-based adoptive cell therapy (ACT) has exhibited excellent antitumoral efficacy exemplified by the clinical breakthrough of chimeric antigen receptor therapy (CAR-T) in hematologic malignancies. It relies on the pool of functional T cells to retain the developmental potential to serially kill targeted cells. However, failure in the continuous supply and persistence of functional T cells has been recognized as a critical barrier to sustainable responses. Conferring stemness on infused T cells, yielding stem cell-like memory T cells (TSCM) characterized by constant self-renewal and multilineage differentiation similar to pluripotent stem cells, is indeed necessary and promising for enhancing T cell function and sustaining antitumor immunity. Therefore, it is crucial to identify TSCM cell induction regulators and acquire more TSCM cells as resource cells during production and after infusion to improve antitumoral efficacy. Recently, four common cytokine receptor γ chain (γc) family cytokines, encompassing interleukin-2 (IL-2), IL-7, IL-15, and IL-21, have been widely used in the development of long-lived adoptively transferred TSCM in vitro. However, challenges, including their non-specific toxicities and off-target effects, have led to substantial efforts for the development of engineered versions to unleash their full potential in the induction and maintenance of T cell stemness in ACT. In this review, we summarize the roles of the four γc family cytokines in the orchestration of adoptively transferred T cell stemness, introduce their engineered versions that modulate TSCM cell formation and demonstrate the potential of their various combinations. Video Abstract.
Collapse
Affiliation(s)
- Mengshi Luo
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjian Gong
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuewen Zhang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huayi Li
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Ma
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinglei Gao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yong Fang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|