1
|
Kay EJ, Zanivan S. The tumor microenvironment is an ecosystem sustained by metabolic interactions. Cell Rep 2025; 44:115432. [PMID: 40088447 DOI: 10.1016/j.celrep.2025.115432] [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: 07/24/2024] [Revised: 12/09/2024] [Accepted: 02/24/2025] [Indexed: 03/17/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) and immune cells make up two major components of the tumor microenvironment (TME), contributing to an ecosystem that can either support or restrain cancer progression. Metabolism is a key regulator of the TME, providing a means for cells to communicate with and influence each other, modulating tumor progression and anti-tumor immunity. Cells of the TME can metabolically interact directly through metabolite secretion and consumption or by influencing other aspects of the TME that, in turn, stimulate metabolic rewiring in target cells. Recent advances in understanding the subtypes and plasticity of cells in the TME both open up new avenues and create challenges for metabolically targeting the TME to hamper tumor growth and improve response to therapy. This perspective explores ways in which the CAF and immune components of the TME could metabolically influence each other, based on current knowledge of their metabolic states, interactions, and subpopulations.
Collapse
Affiliation(s)
- Emily Jane Kay
- Cancer Research UK Scotland Institute, Glasgow G61 1BD, UK.
| | - Sara Zanivan
- Cancer Research UK Scotland Institute, Glasgow G61 1BD, UK; School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
2
|
Bhutani B, Sharma V, Ganguly NK, Rana R. Unravelling the modified T cell receptor through Gen-Next CAR T cell therapy in Glioblastoma: Current status and future challenges. Biomed Pharmacother 2025; 186:117987. [PMID: 40117901 DOI: 10.1016/j.biopha.2025.117987] [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: 12/23/2024] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025] Open
Abstract
PURPOSE Despite current technological advancements in the treatment of glioma, immediate alleviation of symptoms can be catered by therapeutic modalities, including surgery, chemotherapy, and combinatorial radiotherapy that exploit aberrations of glioma. Additionally, a small number of target antigens, their heterogeneity, and immune evasion are the potential reasons for developing targeted therapies. This oncologic milestone has catalyzed interest in developing immunotherapies against Glioblastoma to improve overall survival and cure patients with high-grade glioma. The next-gen CAR-T Cell therapy is one of the effective immunotherapeutic strategies in which autologous T cells have been modified to express receptors against GBM and it modulates cytotoxicity. METHODS In this review article, we examine preclinical and clinical outcomes, and limitations as well as present cutting-edge techniques to improve the function of CAR-T cell therapy and explore the possibility of combination therapy. FINDINGS To date, several CAR T-cell therapies are being evaluated in clinical trials for GBM and other brain malignancies and multiple preclinical studies have demonstrated encouraging outcomes. IMPLICATIONS CAR-T cell therapy represents a promising therapeutic paradigm in the treatment of solid tumors but a few limitations include, the blood-brain barrier (BBB), antigen escape, tumor microenvironment (TME), tumor heterogeneity, and its plasticity that suppresses immune responses weakens the ability of this therapy. Additional investigation is required that can accurately identify the targets and reflect the similar architecture of glioblastoma, thus optimizing the efficiency of CAR-T cell therapy; allowing for the selection of patients most likely to benefit from immuno-based treatments.
Collapse
Affiliation(s)
- Bhavya Bhutani
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Vyoma Sharma
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Nirmal Kumar Ganguly
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Rashmi Rana
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India.
| |
Collapse
|
3
|
Ogulur I, Mitamura Y, Yazici D, Pat Y, Ardicli S, Li M, D'Avino P, Beha C, Babayev H, Zhao B, Zeyneloglu C, Giannelli Viscardi O, Ardicli O, Kiykim A, Garcia-Sanchez A, Lopez JF, Shi LL, Yang M, Schneider SR, Skolnick S, Dhir R, Radzikowska U, Kulkarni AJ, Imam MB, Veen WVD, Sokolowska M, Martin-Fontecha M, Palomares O, Nadeau KC, Akdis M, Akdis CA. Type 2 immunity in allergic diseases. Cell Mol Immunol 2025; 22:211-242. [PMID: 39962262 PMCID: PMC11868591 DOI: 10.1038/s41423-025-01261-2] [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: 09/16/2024] [Accepted: 01/09/2025] [Indexed: 03/01/2025] Open
Abstract
Significant advancements have been made in understanding the cellular and molecular mechanisms of type 2 immunity in allergic diseases such as asthma, allergic rhinitis, chronic rhinosinusitis, eosinophilic esophagitis (EoE), food and drug allergies, and atopic dermatitis (AD). Type 2 immunity has evolved to protect against parasitic diseases and toxins, plays a role in the expulsion of parasites and larvae from inner tissues to the lumen and outside the body, maintains microbe-rich skin and mucosal epithelial barriers and counterbalances the type 1 immune response and its destructive effects. During the development of a type 2 immune response, an innate immune response initiates starting from epithelial cells and innate lymphoid cells (ILCs), including dendritic cells and macrophages, and translates to adaptive T and B-cell immunity, particularly IgE antibody production. Eosinophils, mast cells and basophils have effects on effector functions. Cytokines from ILC2s and CD4+ helper type 2 (Th2) cells, CD8 + T cells, and NK-T cells, along with myeloid cells, including IL-4, IL-5, IL-9, and IL-13, initiate and sustain allergic inflammation via T cell cells, eosinophils, and ILC2s; promote IgE class switching; and open the epithelial barrier. Epithelial cell activation, alarmin release and barrier dysfunction are key in the development of not only allergic diseases but also many other systemic diseases. Recent biologics targeting the pathways and effector functions of IL4/IL13, IL-5, and IgE have shown promising results for almost all ages, although some patients with severe allergic diseases do not respond to these therapies, highlighting the unmet need for a more detailed and personalized approach.
Collapse
Affiliation(s)
- Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Genetics, Faculty of Veterinary Medicine, Bursa Uludag University, Bursa, Turkey
| | - Manru Li
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Paolo D'Avino
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Carina Beha
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Bingjie Zhao
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Can Zeyneloglu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | | | - Ozge Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Division of Food Processing, Milk and Dairy Products Technology Program, Karacabey Vocational School, Bursa Uludag University, Bursa, Turkey
| | - Ayca Kiykim
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Cerrahpasa School of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Asuncion Garcia-Sanchez
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Biomedical and Diagnostic Science, School of Medicine, University of Salamanca, Salamanca, Spain
| | - Juan-Felipe Lopez
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Li-Li Shi
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Minglin Yang
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Stephan R Schneider
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Stephen Skolnick
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Seed Health Inc., Los Angeles, CA, USA
| | - Raja Dhir
- Seed Health Inc., Los Angeles, CA, USA
| | - Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Abhijeet J Kulkarni
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Manal Bel Imam
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Mar Martin-Fontecha
- Departamento de Quimica Organica, Facultad de Optica y Optometria, Complutense University of Madrid, Madrid, Spain
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.
| |
Collapse
|
4
|
Qin D, Lei Y, Shu P, Zhang Y, Loh YH, Wang Y, Li Q. Supercharging CAR-T cells through transcriptional and epigenetic armoring. Theranostics 2025; 15:3345-3367. [PMID: 40093905 PMCID: PMC11905144 DOI: 10.7150/thno.107908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
Inspired by the remarkable success of CAR-T therapy in hematologic malignancies, research is increasingly focused on adapting this treatment for solid tumors. However, CAR-T efficacy remains limited due to its exhaustion and shortened persistence. Transcription factors and epigenetic modifications play pivotal roles in modulating T cell differentiation and functionality, which have been leveraged in numerous strategies to promote the formation of long-lasting memory cells with stem-like properties and supercharging CAR-T performance. This review highlights pivotal transcriptional factors, such as c-Jun and FOXO1, which enhance and sustain T cell effector function, diminishes exhaustion, and epigenetic regulators like TET2 and DNMT3A, whose knockout promotes memory T subsets formation. We explore their interconnections, downstream targets, biological impacts, and the potential application risks of certain candidates, providing a comprehensive theoretical framework for supercharging CAR-T therapies through transcriptional and epigenetic interventions.
Collapse
Affiliation(s)
- Diyuan Qin
- Cancer Center, Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A ∗ STAR), Singapore 138673, Singapore
| | - Yanna Lei
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Pei Shu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yugu Zhang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuin-Han Loh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A ∗ STAR), Singapore 138673, Singapore
| | - Yongsheng Wang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Cancer Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qijing Li
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A ∗ STAR), Singapore 138673, Singapore
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
| |
Collapse
|
5
|
Zhao J, Wang K, Li Y, Hu X, Liu H, Zhao J, Li L, Yuan S. Impact of radiation dose to immune cells on survival in patients with esophageal cancer receiving neoadjuvant chemoradiotherapy: a retrospective analysis. BMC Cancer 2025; 25:238. [PMID: 39934708 DOI: 10.1186/s12885-025-13602-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 01/28/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Neoadjuvant chemoradiotherapy (NCRT) is the standard treatment for locally advanced esophageal squamous cell carcinoma (ESCC). This study aims to explore the effect of the estimated dose of radiation to immune cells (EDRIC) on lymphocytes in patients with locally advanced ESCC undergoing NCRT and evaluate the impact of EDRIC on patient prognosis. METHODS This retrospective study included patients with locally advanced ESCC who received NCRT followed by radical esophagectomy at our institution between March 2016 and December 2022. EDRIC was calculated as a function of the number of fractions and the average doses to the lungs, heart, and remaining body. The optimal cut-off value of EDRIC was determined based on disease-free survival (DFS), and patients were divided into two groups according to this cut-off value. Kaplan-Meier and Cox proportional hazards regression analyses were used to estimate overall survival (OS) and DFS. RESULTS The 182 patients included in the study had a mean EDRIC of 5.41 ± 1.70 Gy and an EDRIC cut-off value of 6.86 Gy. Patients with an EDRIC ≤ 6.86 Gy had a significant advantage in DFS (hazard ratio [HR]: 0.37, 95% confidence interval [CI]: 0.20-0.68, P = 0.001) and OS (HR: 0.22, 95% CI: 0.10-0.48, P = 0.001). EDRIC was closely associated with the lymphocyte nadir (P < 0.001) and lymphocytopenia values (P < 0.001). Patients with a lymphocyte nadir ≤ 0.16 × 109/L had poorer DFS (HR: 2.04, 95% CI: 1.07-3.88, P = 0.025) and OS (HR: 2.52, 95% CI: 1.14-5.56, P = 0.018). CONCLUSION EDRIC is an independent prognostic factor for patients with locally advanced ESCC undergoing NCRT. A higher EDRIC is associated with poorer DFS and OS and is closely related to reduced lymphocyte counts. As a quantifiable parameter, future research should explore methods of lowering EDRIC during NCRT while ensuring adequate tumor coverage and sufficient dosage.
Collapse
Affiliation(s)
- Junfeng Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, 250117, Shandong, China
| | - Kang Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, 250117, Shandong, China
| | - Ying Li
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xinying Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, 250117, Shandong, China
| | - Haoyu Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, 250117, Shandong, China
| | - Jingjing Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, 250117, Shandong, China
| | - Li Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, 250117, Shandong, China
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 17 Lujiang Road, Hefei, 230001, Anhui, China
| | - Shuanghu Yuan
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, 250117, Shandong, China.
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 17 Lujiang Road, Hefei, 230001, Anhui, China.
| |
Collapse
|
6
|
Wang B, Xia Y, Zhou C, Zeng Y, Son HG, Demehri S. CD4+ T helper 2 cell-macrophage crosstalk induces IL-24-mediated breast cancer suppression. JCI Insight 2025; 10:e180962. [PMID: 39782693 PMCID: PMC11721301 DOI: 10.1172/jci.insight.180962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 11/08/2024] [Indexed: 01/12/2025] Open
Abstract
CD4+ T cells contribute to antitumor immunity and are implicated in the efficacy of cancer immunotherapies. In particular, CD4+ T helper 2 (Th2) cells were recently found to block spontaneous breast carcinogenesis. However, the antitumor potential of Th2 cells in targeting established breast cancer remains uncertain. Herein, we demonstrate that Th2 cells induced by the topical calcipotriol/thymic stromal lymphopoietin cytokine axis suppressed the growth of established mammary tumors in mice. Interleukin-24 (IL-24), an anticancer cytokine, was highly upregulated in macrophages infiltrating calcipotriol-treated mammary tumors. Macrophages expressed IL-24 in response to IL-4 signaling in combination with Toll-like receptor 4 (TLR4) agonists (e.g., HMGB1) in vitro. Calcipotriol treatment significantly increased HMGB1 release by tumor cells in vivo. CD4+ T cell depletion reduced HMGB1 and IL-24 expression, reversing calcipotriol's therapeutic efficacy. Macrophage depletion and TLR4 inhibition also reduced the therapeutic efficacy of calcipotriol. Importantly, calcipotriol treatment failed to control mammary tumors lacking the IL-24 receptor on tumor cells. Collectively, our findings reveal that Th2 cell-macrophage crosstalk leads to IL-24-mediated tumor cell death, highlighting a promising therapeutic strategy to tackle breast cancer.
Collapse
Affiliation(s)
- Bo Wang
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Urology and
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yun Xia
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Can Zhou
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Yuhan Zeng
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Heehwa G. Son
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Shadmehr Demehri
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
7
|
Wagner M, Nishikawa H, Koyasu S. Reinventing type 2 immunity in cancer. Nature 2025; 637:296-303. [PMID: 39780006 DOI: 10.1038/s41586-024-08194-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 10/10/2024] [Indexed: 01/11/2025]
Abstract
Our understanding of type 2 immunity has undergone a substantial transformation in recent years, revealing previously unknown functions. Beyond its canonical role in defence against parasitic helminth infections, type 2 immunity safeguards the host through additional mechanisms, including the suppression of excessive type 1 immune responses, regulation of tissue repair and maintenance of adipose tissue homeostasis. However, unlike type 1 immune responses, type 2 immunity is perceived as a potential promoter of tumorigenesis. Emerging evidence challenges this perspective, painting a more nuanced picture in which type 2 immunity might protect against or even actively suppress tumour growth and progression. In this Review, we explore discoveries that highlight the potential of type 2 immunity in reshaping the landscape of cancer immunotherapies.
Collapse
Affiliation(s)
- Marek Wagner
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network-PORT Polish Center for Technology Development, Wrocław, Poland.
| | - Hiroyoshi Nishikawa
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Cancer Immunology, Research Institute/EPOC, National Cancer Center, Tokyo, Japan
| | - Shigeo Koyasu
- Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- National Institutes for Quantum Science and Technology (QST), Chiba, Japan.
| |
Collapse
|
8
|
Biernacki MA, Bleakley M. Clinical trials, challenges, and changes in TCR-based therapeutics for hematologic malignancies. Expert Rev Hematol 2025; 18:21-31. [PMID: 39667756 DOI: 10.1080/17474086.2024.2441962] [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: 03/15/2024] [Revised: 08/14/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION T cells engineered to express antigen-specific T cell receptors (TCR; TCR-T) are a promising class of immunotherapeutic for patients with hematologic malignancies. Like chimeric antigen receptor-engineered T cells (CAR-T), TCR-T are cell products with defined specificity and composition. Unlike CAR-T, TCR-T can recognize targets arising both from intracellular and cell surface proteins and leverage the sensitivity of natural TCR signaling machinery. A growing number of TCR-T targeting various antigens in different hematologic malignancies are in early-phase clinical trials, and more are in preclinical development. AREAS COVERED This review covers results from early-phase TCR-T clinical trials for hematologic malignancies. Challenges in the field are reviewed, including identifying optimal targets, engaging CD4+ help for CD8+ T cells, and overcoming tumor-induced suppression; recent innovations to overcome these challenges are also highlighted. EXPERT OPINION In the future, TCR-T's promise for hematologic malignancies will be borne out in later-phase clinical trials and approvals for clinical use. Improved antigen discovery methods will help build the toolbox of targets needed for broadly applicable TCR-T. Rationally designed TCR-T modifications including incorporation of accessory receptors and gene editing will enhance TCR-T function. New hybrid receptors combining features of TCR and CAR will enter the clinic.
Collapse
Affiliation(s)
| | - Marie Bleakley
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| |
Collapse
|
9
|
Zhu L, Cui X, Yan Z, Tao Y, Shi L, Zhang X, Yao Y, Shi L. Design and evaluation of a multi-epitope DNA vaccine against HPV16. Hum Vaccin Immunother 2024; 20:2352908. [PMID: 38780076 PMCID: PMC11123455 DOI: 10.1080/21645515.2024.2352908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024] Open
Abstract
Cervical cancer, among the deadliest cancers affecting women globally, primarily arises from persistent infection with high-risk human papillomavirus (HPV). To effectively combat persistent infection and prevent the progression of precancerous lesions into malignancy, a therapeutic HPV vaccine is under development. This study utilized an immunoinformatics approach to predict epitopes of cytotoxic T lymphocytes (CTLs) and helper T lymphocytes (HTLs) using the E6 and E7 oncoproteins of the HPV16 strain as target antigens. Subsequently, through meticulous selection of T-cell epitopes and other necessary elements, a multi-epitope vaccine was constructed, exhibiting good immunogenic, physicochemical, and structural characteristics. Furthermore, in silico simulations showed that the vaccine not only interacted well with toll-like receptors (TLR2/TLR3/TLR4), but also induced a strong innate and adaptive immune response characterized by elevated Th1-type cytokines, such as interferon-gamma (IFN-γ) and interleukin-2 (IL2). Additionally, our study investigated the effects of different immunization intervals on immune responses, aiming to optimize a time-efficient immunization program. In animal model experiments, the vaccine exhibited robust immunogenic, therapeutic, and prophylactic effects. Administered thrice, it consistently induced the expansion of specific CD4 and CD8 T cells, resulting in substantial cytokines release and increased proliferation of memory T cell subsets in splenic cells. Overall, our findings support the potential of this multi-epitope vaccine in combating HPV16 infection and signify its candidacy for future HPV vaccine development.
Collapse
Affiliation(s)
- Lanfang Zhu
- Department of Immunogenetics, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, China
| | - Xiangjie Cui
- Department of Immunogenetics, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, China
| | - Zhiling Yan
- Department of Gynaecologic Oncology, The No. 3 Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yufen Tao
- Department of Immunogenetics, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, China
| | - Lei Shi
- Department of Immunogenetics, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, China
| | - Xinwen Zhang
- Department of Immunogenetics, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, China
| | - Yufeng Yao
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, China
| | - Li Shi
- Department of Immunogenetics, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, China
| |
Collapse
|
10
|
Luo D, Zhong Q, Yue H, Wang J, Liang Q, Liu W, Zhu X. The predictors of lymphopenia and its effects on survival in locally advanced esophageal squamous cell carcinoma. Cancer Biol Ther 2024; 25:2371632. [PMID: 38946404 PMCID: PMC11218796 DOI: 10.1080/15384047.2024.2371632] [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: 12/15/2022] [Accepted: 06/19/2024] [Indexed: 07/02/2024] Open
Abstract
To investigate the impact of the effective radiation dose to immune cells (EDIC) and gross tumor volume (GTV) on lymphopenia and survival in patients with locally advanced esophageal squamous cell carcinoma (LAESCC). Between January 2013 and December 2020, 272 LAESCC patients were treated with definitive radiotherapy in two institutions. Based on radiation doses to the lungs, heart, and body region scanned, EDIC was calculated as an equal uniform dose to the total blood considering blood flow and fraction effect. The radiotherapy plan was used to calculate the GTVs. Lymphopenia was graded based on the lowest lymphocyte count during RT. The overall survival (OS), progress-free survival (PFS), and local recurrence-free survival (LRFS) were analyzed statistically. The lowest lymphocyte count was significantly correlated with EDIC (r= -0.389, p < .001) and GTV (r= -0.211, p < .001). Lymphopenia, EDIC, and GTV are risk factors for patients with ESCC. In a Kaplan-Meier analysis with EDIC and GTV as stratification factors, lymphopenia was not associated with OS in the EDIC>12.9 Gy group (p = .294)and EDIC ≤ 12.9 Gy group, and it was also not associated with OS in GTV>68.8 cm3 group (p = .242) and GTV ≤ 68.8 cm3 group(p = .165). GTV and EDIC had an impact on the relationship between lymphopenia and OS in patients with LAESCC undergoing definitive RT. Poorer OS, PFS, and LRFS are correlated with lymphopenia, higher EDIC, and larger GTV.
Collapse
Affiliation(s)
- Danjing Luo
- Department of Radiation Oncology, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qiulu Zhong
- Department of Radiation Oncology, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Haiying Yue
- Department of Radiation Oncology, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jue Wang
- Department of Radiation Oncology, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qianfu Liang
- Department of Radiation Oncology, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wenqi Liu
- Department of Radiation Oncology, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaodong Zhu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
- Department of Oncology, Wuming Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
11
|
Schol P, van Elsas MJ, Middelburg J, Nijen Twilhaar MK, van Hall T, van der Sluis TC, van der Burg SH. Myeloid effector cells in cancer. Cancer Cell 2024; 42:1997-2014. [PMID: 39658540 DOI: 10.1016/j.ccell.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/21/2024] [Accepted: 11/06/2024] [Indexed: 12/12/2024]
Abstract
The role of myeloid cells in tumor immunity is multifaceted. While dendritic cells support T cell-mediated tumor control, the highly heterogenous populations of macrophages, neutrophils, and immature myeloid cells were generally considered immunosuppressive. This view has led to effective therapies reinvigorating tumor-reactive T cells; however, targeting the immunosuppressive effects of macrophages and neutrophils to boost the cancer immunity cycle was clinically less successful. Recent studies interrogating the role of immune cells in the context of successful immunotherapy affirm the key role of T cells, but simultaneously challenge the idea that the cytotoxic function of T cells is the main contributor to therapy-driven tumor regression. Rather, therapy-activated intra-tumoral T cells recruit and activate or reprogram several myeloid effector cell types, the presence of which is necessary for tumor rejection. Here, we reappreciate the key role of myeloid effector cells in tumor rejection as this may help to shape future successful immunotherapies.
Collapse
Affiliation(s)
- Pieter Schol
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Marit J van Elsas
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Jim Middelburg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Maarten K Nijen Twilhaar
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Tetje C van der Sluis
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
12
|
Tao Z, Chyra Z, Kotulová J, Celichowski P, Mihályová J, Charvátová S, Hájek R. Impact of T cell characteristics on CAR-T cell therapy in hematological malignancies. Blood Cancer J 2024; 14:213. [PMID: 39627220 PMCID: PMC11615218 DOI: 10.1038/s41408-024-01193-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 12/06/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment paradigms for hematological malignancies. However, more than half of these patients cannot achieve sustainable tumor control, partially due to the inadequate potency of CAR-T cells in eradicating tumor cells. T cells are crucial components of the anti-tumor immune response, and multiple intrinsic T-cell features significantly influence the outcomes of CAR-T cell therapy. Herein, we review progressing research on T-cell characteristics that impact the effectiveness of CAR-T cells, including T-cell exhaustion, memory subsets, senescence, regulatory T-cells, the CD4+ to CD8+ T-cell ratio, metabolism, and the T-cell receptor repertoire. With comprehensive insight into the biological processes underlying successful CAR-T cell therapy, we will further refine the applications of these novel therapeutic modalities, and enhance their efficacy and safety for patients.
Collapse
Affiliation(s)
- Zhongfei Tao
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Zuzana Chyra
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Jana Kotulová
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Piotr Celichowski
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Jana Mihályová
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Sandra Charvátová
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Roman Hájek
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic.
| |
Collapse
|
13
|
Park DH, Bhojnagarwala PS, Liaw K, Bordoloi D, Tursi NJ, Zhao S, Binder ZA, O’Rourke D, Weiner DB. Novel tri-specific T-cell engager targeting IL-13Rα2 and EGFRvIII provides long-term survival in heterogeneous GBM challenge and promotes antitumor cytotoxicity with patient immune cells. J Immunother Cancer 2024; 12:e009604. [PMID: 39622583 PMCID: PMC11624777 DOI: 10.1136/jitc-2024-009604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is known for its high antigenic heterogeneity, which undermines the effectiveness of monospecific immunotherapies. Multivalent immunotherapeutic strategies that target multiple tumor antigens simultaneously could enhance clinical outcomes by preventing antigen-driven tumor escape mechanisms. METHODS We describe novel trivalent antibodies, DNA-encoded tri-specific T-cell engagers (DTriTEs), targeting two GBM antigens, epidermal growth factor receptor variant III (EGFRvIII) and IL-13Rα2, and engaging T cells through CD3. We engineered three DTriTE constructs, each with a unique arrangement of the antigen-binding fragments within a single-chain sequence. We assessed the binding efficiency and cytotoxic activity of these DTriTEs in vitro on target cells expressing relevant antigens. In vivo efficacy was tested in immunocompromised mice, including a longitudinal expression study post-administration and a survival analysis in an NOD scid gamma (NSG)-K mouse model under a heterogeneous tumor burden. RNA sequencing of DTriTE-activated T cells was employed to identify the molecular pathways influenced by the treatment. The antitumor cytotoxicity of patient-derived immune cells was evaluated following stimulation by DTriTE to assess its potential effectiveness in a clinical setting. RESULTS All DTriTE constructs demonstrated strong binding to EGFRvIII and IL-13Rα2-expressing cells, induced significant T cell-mediated cytotoxicity, and enhanced cytokine production (interferon-γ, tumor necrosis factor (TNF)-α, and interleukin(IL)-2). The lead construct, DT2035, sustained expression for over 105 days in vivo and exhibited elimination of tumor burden in a heterogeneous intracranial GBM model, outperforming monospecific antibody controls. In extended survival studies using the NSG-K model, DT2035 achieved a 67% survival rate over 120 days. RNA sequencing of DTriTE-activated T cells showed that DT2035 enhances genes linked to cytotoxicity, proliferation, and immunomodulation, reflecting potent immune activation. Finally, DT2035 effectively induced target-specific cytotoxicity in post-treatment peripheral blood mononuclear cells from patients with GBM, highlighting its potential for clinical effectiveness. CONCLUSIONS DTriTEs exhibit potent anti-tumor effects and durable in vivo activity, offering promising therapeutic potential against GBM. These findings support further development of such multivalent therapeutic strategies to improve treatment outcomes in GBM and potentially other antigenically heterogeneous tumors. The opportunity to advance such important therapies either through biologic delivery or direct in vivo nucleic acid production is compelling.
Collapse
Affiliation(s)
- Daniel H Park
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Kevin Liaw
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Devivasha Bordoloi
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Nicholas J Tursi
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Shushu Zhao
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Zev A Binder
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- GBM Translational Center of Excellence, Abramson Cancer Center, Philadelphia, Pennsylvania, USA
| | - Donald O’Rourke
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- GBM Translational Center of Excellence, Abramson Cancer Center, Philadelphia, Pennsylvania, USA
| | - David B Weiner
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
Arnold IC, Munitz A. Spatial adaptation of eosinophils and their emerging roles in homeostasis, infection and disease. Nat Rev Immunol 2024; 24:858-877. [PMID: 38982311 DOI: 10.1038/s41577-024-01048-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 07/11/2024]
Abstract
Eosinophils are bone marrow-derived granulocytes that are traditionally associated with type 2 immune responses, such as those that occur during parasite infections and allergy. Emerging evidence demonstrates the remarkable functional plasticity of this elusive cell type and its pleiotropic functions in diverse settings. Eosinophils broadly contribute to tissue homeostasis, host defence and immune regulation, predominantly at mucosal sites. The scope of their activities primarily reflects the breadth of their portfolio of secreted mediators, which range from cytotoxic cationic proteins and reactive oxygen species to multiple cytokines, chemokines and lipid mediators. Here, we comprehensively review basic eosinophil biology that is directly related to their activities in homeostasis, protective immunity, regeneration and cancer. We examine how dysregulation of these functions contributes to the physiopathology of a broad range of inflammatory diseases. Furthermore, we discuss recent findings regarding the tissue compartmentalization and adaptation of eosinophils, shedding light on the factors that likely drive their functional diversification within tissues.
Collapse
Affiliation(s)
- Isabelle C Arnold
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland.
| | - Ariel Munitz
- Department of Clinical Microbiology and Immunology, Faculty of Medical and Health Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel.
| |
Collapse
|
15
|
Pollack IF, Felker J, Frederico SC, Raphael I, Kohanbash G. Immunotherapy for pediatric low-grade gliomas. Childs Nerv Syst 2024; 40:3263-3275. [PMID: 38884777 DOI: 10.1007/s00381-024-06491-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/01/2024] [Indexed: 06/18/2024]
Abstract
Pediatric low-grade gliomas (pLGGs) are the most common brain tumor types affecting children. Although gross-total resection remains the treatment of choice, many tumors are not amenable to complete removal, because they either involve midline structures, such as the optic chiasm or hypothalamus, and are not conducive to aggressive resection, or have diffuse biological features and blend with the surrounding brain. Historically, radiation therapy was used as the second-line option for disease control, but with the recognition that this often led to adverse long-term sequelae, particularly in young children, conventional chemotherapy assumed a greater role in initial therapy for unresectable tumors. A variety of agents demonstrated activity, but long-term disease control was suboptimal, with more than 50% of tumors exhibiting disease progression within 5 years. More recently, it has been recognized that a high percentage of these tumors in children exhibit constitutive activation of the mitogen-activated protein kinase (MAPK) pathway because of BRAF translocations or mutations, NFI mutations, or a host of other anomalies that converged on MAPK. This led to phase 1, 2, and 3 trials that explored the activity of blocking this signaling pathway, and the efficacy of this approach compared to conventional chemotherapy. Despite initial promise of these strategies, not all children tolerate this therapy, and many tumors resume growth once MAPK inhibition is stopped, raising concern that long-term and potentially life-long treatment will be required to maintain tumor control, even among responders. This observation has led to interest in other treatments, such as immunotherapy, that may delay or avoid the need for additional treatments. This chapter will summarize the place of immunotherapy in the current armamentarium for these tumors and discuss prior results and future options to improve disease control, with a focus on our prior efforts and experience in this field.
Collapse
Affiliation(s)
- Ian F Pollack
- Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Department of Neurosurgery, UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.
| | - James Felker
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurosurgery, UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Stephen C Frederico
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurosurgery, UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Itay Raphael
- Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurosurgery, UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Gary Kohanbash
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurosurgery, UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| |
Collapse
|
16
|
Deo AS, Shrijana, S U S, Karun S, Bisaria K, Sarkar K. Participation of T cells in generating immune protection against cancers. Pathol Res Pract 2024; 262:155534. [PMID: 39180801 DOI: 10.1016/j.prp.2024.155534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 08/09/2024] [Accepted: 08/11/2024] [Indexed: 08/27/2024]
Abstract
T cells are essential to the immune system's reaction. The major job of the immune system is to identify and get rid of any abnormal or malignant cells in the body. White blood cells called T cells coordinate and carry out immunological responses, including identifying and eliminating cancer cells. It mostly consists of two types called helper T-cells and cytotoxic T-cells. Together, they create an efficient reaction against cancer. Both the primary T cell subtype - CD4+ and CD8+ Tcells have specific role to play in our immune system.CD4+ T cells are limited to MHC-II molecules and acts as helper cell by activating and enhancing other immune cells. On the other side CD8+ T cells are called the killer cells as they eradicate the abnormal and contaminated cells and are limited to MHC-I molecules. The malignant cells are destroyed when cytotoxic T cells come into direct contact with them. This happens via number of processes, including TCR recognition, the release of cytotoxic chemicals, and finally the activation of the immune system. T cell receptors on the surface of cytotoxic T cells allow them to identify tumour cells and these T cells release harmful chemicals like perforins and granzymes when they connect to malignant cells. T-cells that have been stimulated release cytokines such as gamma interferon. T-cells can also acquire memory responses that improve their capacity for recognition and response. Helper T-cells contribute to the development of an immune response. It entails coordination and activation as well as the enlistment of additional immune cells, including macrophages and natural killer cells, to assist in the eradication of cancer cells. Despite the fact that the cancer frequently creates defence systems to circumvent their immune response. Together, these activities support the immune surveillance and T-cell-mediated regulation of cancer cells. Treatments like chemotherapy, radiation, and surgery are main ways to treat cancer but immunotherapy has been emerging since last few decades. These immune specific treatments have shown huge positive result. CAR T cell therapy is a promising weapon to fight again blood cancer and it works by focusing on our immune system to fight and eliminate cancer.
Collapse
Affiliation(s)
- Anisha Singha Deo
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Shrijana
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Sruthika S U
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Shreya Karun
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Kashish Bisaria
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Koustav Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India.
| |
Collapse
|
17
|
Lee H, Bae AN, Yang H, Lee JH, Park JH. Modulation of PRC1 Promotes Anticancer Effects in Pancreatic Cancer. Cancers (Basel) 2024; 16:3310. [PMID: 39409930 PMCID: PMC11475828 DOI: 10.3390/cancers16193310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/11/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Pancreatic cancer, while relatively uncommon, is extrapolated to become the second leading cause of cancer-related deaths worldwide. Despite identifying well-known markers like the KRAS gene, the exact regulation of pancreatic cancer progression remains elusive. Methods: Clinical value of PRC1 was analyzed using bioinformatics database. The role of PRC1 was further evaluated through cell-based assays, including viability, wound healing, and sensitivity with the drug. Results: We demonstrate that PRC1 was significantly overexpressed in pancreatic cancer compared to pancreases without cancer, as revealed through human databases and cell lines analysis. Furthermore, high PRC1 expression had a negative correlation with CD4+ T cells, which are crucial for the immune response against cancers. Additionally, PRC1 showed a positive correlation with established pancreatic cancer markers. Silencing PRC1 expression using siRNA significantly inhibited cancer cell proliferation and viability and increased chemotherapeutic drug sensitivity. Conclusions: These findings suggest that targeting PRC1 in pancreatic cancer may enhance immune cell infiltration and inhibit cancer cell proliferation, offering a promising avenue for developing anticancer therapies.
Collapse
Affiliation(s)
| | | | | | | | - Jong Ho Park
- Department of Anatomy, School of Medicine, Keimyung University, Daegu 42601, Republic of Korea
| |
Collapse
|
18
|
Krakow EF, Brault M, Summers C, Cunningham TM, Biernacki MA, Black RG, Woodward KB, Vartanian N, Kanaan SB, Yeh AC, Dossa RG, Bar M, Cassaday RD, Dahlberg A, Till BG, Denker AE, Yeung CCS, Gooley TA, Maloney DG, Riddell SR, Greenberg PD, Chapuis AG, Newell EW, Furlan SN, Bleakley M. HA-1-targeted T-cell receptor T-cell therapy for recurrent leukemia after hematopoietic stem cell transplantation. Blood 2024; 144:1069-1082. [PMID: 38683966 PMCID: PMC11406181 DOI: 10.1182/blood.2024024105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/27/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
ABSTRACT Relapse is the leading cause of death after allogeneic hematopoietic stem cell transplantation (HCT) for leukemia. T cells engineered by gene transfer to express T cell receptors (TCR; TCR-T) specific for hematopoietic-restricted minor histocompatibility (H) antigens may provide a potent selective antileukemic effect post-HCT. We conducted a phase 1 clinical trial using a novel TCR-T product targeting the minor H antigen, HA-1, to treat or consolidate treatment of persistent or recurrent leukemia and myeloid neoplasms. The primary objective was to evaluate the feasibility and safety of administration of HA-1 TCR-T after HCT. CD8+ and CD4+ T cells expressing the HA-1 TCR and a CD8 coreceptor were successfully manufactured from HA-1-disparate HCT donors. One or more infusions of HA-1 TCR-T following lymphodepleting chemotherapy were administered to 9 HCT recipients who had developed disease recurrence after HCT. TCR-T cells expanded and persisted in vivo after adoptive transfer. No dose-limiting toxicities occurred. Although the study was not designed to assess efficacy, 4 patients achieved or maintained complete remissions following lymphodepletion and HA-1 TCR-T, with 1 patient still in remission at >2 years. Single-cell RNA sequencing of relapsing/progressive leukemia after TCR-T therapy identified upregulated molecules associated with T-cell dysfunction or cancer cell survival. HA-1 TCR-T therapy appears feasible and safe and shows preliminary signals of efficacy. This clinical trial was registered at ClinicalTrials.gov as #NCT03326921.
Collapse
Affiliation(s)
- Elizabeth F. Krakow
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Michelle Brault
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Corinne Summers
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Cancer and Blood Disorders Center, Seattle Children's Hospital, Seattle, WA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA
| | - Tanya M. Cunningham
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Melinda A. Biernacki
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - R. Graeme Black
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Kyle B. Woodward
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Nicole Vartanian
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Sami B. Kanaan
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Albert C. Yeh
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Robson G. Dossa
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Merav Bar
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Ryan D. Cassaday
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Ann Dahlberg
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Cancer and Blood Disorders Center, Seattle Children's Hospital, Seattle, WA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA
| | - Brian G. Till
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | | - Cecilia C. S. Yeung
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA
| | - Ted A. Gooley
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - David G. Maloney
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Stanley R. Riddell
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Philip D. Greenberg
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Immunology, University of Washington School of Medicine, Seattle, WA
| | - Aude G. Chapuis
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Evan W. Newell
- Vaccine and Infection Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Scott N. Furlan
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Cancer and Blood Disorders Center, Seattle Children's Hospital, Seattle, WA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA
| | - Marie Bleakley
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Cancer and Blood Disorders Center, Seattle Children's Hospital, Seattle, WA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
19
|
Cánovas-Cervera I, Nacher-Sendra E, Suay G, Lahoz A, García-Giménez JL, Mena-Mollá S. Role of miRNAs as epigenetic regulators of immune checkpoints in lung cancer immunity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 390:109-139. [PMID: 39864893 DOI: 10.1016/bs.ircmb.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The advent of immunotherapy in cancer has provided new avenues in the treatment of many malignancies at various stages. Specifically, immune checkpoint inhibitors (ICIs) have transformed the field of lung cancer treatment. However, since some tumors can evade the immune system, not all patients respond properly. Recent research has provided evidence showing how microRNAs (miRNAs) are involved in regulating many immune checkpoints. MiRNAs have demonstrated their ability to modulate immune evasion of tumor cells. Currently, reliable markers are being sought to predict the efficacy of immunotherapy in these types of cancers. Therefore, the association of serum miRNAs and the response of ICIs in lung cancer is under study. Many miRNA molecules and their corresponding target genes have been identified in the regulation of chemoresistance. Therefore, elucidating how these miRNAs control the function of immune checkpoints, as well as the effectiveness of therapies based on ICIs set the basis for the development of new biomarkers to predict treatment response to ICIs. This chapter delves into the molecular role of miRNAs interacting with ICs, such as PD-1 and PD-L1, and the clinical utility of miRNAs, such as miR-16, miR-146a, and miR-335, in predicting treatment response to ICI-based therapy in lung cancer. The aim is to provide a deep insight of the current landscape, serving as a cornerstone for further research.
Collapse
Affiliation(s)
- Irene Cánovas-Cervera
- INCLIVA Health Research Institute, INCLIVA, Valencia, Spain; Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | - Elena Nacher-Sendra
- INCLIVA Health Research Institute, INCLIVA, Valencia, Spain; Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | - Guillermo Suay
- Medical Oncology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Agustin Lahoz
- Biomarkers and Precision Medicine Unit, Health Research Institute-Hospital La Fe, Valencia, Spain; Analytical Unit, Health Research Institute-Hospital La Fe, Valencia, Spain
| | - José Luis García-Giménez
- INCLIVA Health Research Institute, INCLIVA, Valencia, Spain; Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain; Consortium Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, Valencia, Spain.
| | - Salvador Mena-Mollá
- INCLIVA Health Research Institute, INCLIVA, Valencia, Spain; Department of Physiology, Faculty of Pharmacy, University of Valencia, Burjassot, Spain
| |
Collapse
|
20
|
Liu J, He C, Tan W, Zheng JH. Path to bacteriotherapy: From bacterial engineering to therapeutic perspectives. Life Sci 2024; 352:122897. [PMID: 38971366 DOI: 10.1016/j.lfs.2024.122897] [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: 01/11/2024] [Revised: 06/30/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
The major reason for the failure of conventional therapies is the heterogeneity and complexity of tumor microenvironments (TMEs). Many malignant tumors reprogram their surface antigens to evade the immune surveillance, leading to reduced antigen-presenting cells and hindered T-cell activation. Bacteria-mediated cancer immunotherapy has been extensively investigated in recent years. Scientists have ingeniously modified bacteria using synthetic biology and nanotechnology to enhance their biosafety with high tumor specificity, resulting in robust anticancer immune responses. To enhance the antitumor efficacy, therapeutic proteins, cytokines, nanoparticles, and chemotherapeutic drugs have been efficiently delivered using engineered bacteria. This review provides a comprehensive understanding of oncolytic bacterial therapies, covering bacterial design and the intricate interactions within TMEs. Additionally, it offers an in-depth comparison of the current techniques used for bacterial modification, both internally and externally, to maximize their therapeutic effectiveness. Finally, we outlined the challenges and opportunities ahead in the clinical application of oncolytic bacterial therapies.
Collapse
Affiliation(s)
- Jinling Liu
- The Affiliated Xiangtan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha 410082, China; College of Biology, Hunan University, Changsha 410082, China
| | - Chongsheng He
- College of Biology, Hunan University, Changsha 410082, China
| | - Wenzhi Tan
- School of Food Science and Bioengineering, Changsha University of Science & Technology, Changsha, Hunan 410114, China.
| | - Jin Hai Zheng
- The Affiliated Xiangtan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha 410082, China.
| |
Collapse
|
21
|
Espinosa-Carrasco G, Chiu E, Scrivo A, Zumbo P, Dave A, Betel D, Kang SW, Jang HJ, Hellmann MD, Burt BM, Lee HS, Schietinger A. Intratumoral immune triads are required for immunotherapy-mediated elimination of solid tumors. Cancer Cell 2024; 42:1202-1216.e8. [PMID: 38906155 PMCID: PMC11413804 DOI: 10.1016/j.ccell.2024.05.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 03/11/2024] [Accepted: 05/29/2024] [Indexed: 06/23/2024]
Abstract
Tumor-specific CD8+ T cells are frequently dysfunctional and unable to halt tumor growth. We investigated whether tumor-specific CD4+ T cells can be enlisted to overcome CD8+ T cell dysfunction within tumors. We find that the spatial positioning and interactions of CD8+ and CD4+ T cells, but not their numbers, dictate anti-tumor responses in the context of adoptive T cell therapy as well as immune checkpoint blockade (ICB): CD4+ T cells must engage with CD8+ T cells on the same dendritic cell during the effector phase, forming a three-cell-type cluster (triad) to license CD8+ T cell cytotoxicity and cancer cell elimination. When intratumoral triad formation is disrupted, tumors progress despite equal numbers of tumor-specific CD8+ and CD4+ T cells. In patients with pleural mesothelioma treated with ICB, triads are associated with clinical responses. Thus, CD4+ T cells and triads are required for CD8+ T cell cytotoxicity during the effector phase and tumor elimination.
Collapse
Affiliation(s)
| | - Edison Chiu
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Aurora Scrivo
- Department of Developmental and Molecular Biology, and Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Paul Zumbo
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA; Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
| | - Asim Dave
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Doron Betel
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA; Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sung Wook Kang
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Hee-Jin Jang
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Matthew D Hellmann
- Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Bryan M Burt
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA; Division of Thoracic Surgery, University of California Los Angeles, Los Angeles, CA, USA
| | - Hyun-Sung Lee
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Andrea Schietinger
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
22
|
Zhang K, Zhu J, Wang P, Chen Y, Wang Z, Ge X, Wu J, Chen L, Lu Y, Xu P, Yao J. Plasma metabolites as mediators in immune cell-pancreatic cancer risk: insights from Mendelian randomization. Front Immunol 2024; 15:1402113. [PMID: 38933268 PMCID: PMC11199692 DOI: 10.3389/fimmu.2024.1402113] [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: 03/16/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Background Immune cells play a crucial role in the development and progression of pancreatic cancer, yet the causal relationship remains uncertain due to complex immune microenvironments and conflicting research findings. Mendelian randomization (MR), this study aims to delineate the causal relationships between immune cells and pancreatic cancer while identifying intermediary factors. Methods The genome-wide association study (GWAS) data on immune cells, pancreatic cancer, and plasma metabolites are derived from public databases. In this investigation, inverse variance weighting (IVW) as the primary analytical approach to investigate the causal relationship between exposure and outcome. Furthermore, this study incorporates MR-Egger, simple mode, weighted median, and weighted mode as supplementary analytical approaches. To ensure the reliability of our findings, we further assessed horizontal pleiotropy and heterogeneity and evaluated the stability of MR results using the Leave-one-out method. In conclusion, this study employed mediation analysis to elucidate the potential mediating effects of plasma metabolites. Results Our investigation revealed a causal relationship between immune cells and pancreatic cancer, highlighting the pivotal roles of CD11c+ monocytes (odds ratio, ORIVW=1.105; 95% confidence interval, 95%CI: 1.002-1.218; P=0.045), HLA DR+ CD4+ antigen-presenting cells (ORIVW=0.920; 95%CI: 0.873-0.968; P=0.001), and HLA DR+ CD8br T cells (ORIVW=1.058; 95%CI: 1.002-1.117; P=0.041) in pancreatic cancer progression. Further mediation analysis indicated that oxalate (proportion of mediation effect in total effect: -11.6%, 95% CI: -89.7%, 66.6%) and the mannose to trans-4-hydroxyproline ratio (-19.4, 95% CI: -136%, 96.8%) partially mediate the relationship between HLA DR+ CD8br T cells and pancreatic cancer in nature. In addition, our analysis indicates that adrenate (-8.39%, 95% CI: -18.3%, 1.54%) plays a partial mediating role in the association between CD11c+ monocyte and pancreatic cancer, while cortisone (-26.6%, 95% CI: 138%, -84.8%) acts as a partial mediator between HLA DR+ CD4+ AC and pancreatic cancer. Conclusion This MR investigation provides evidence supporting the causal relationship between immune cell and pancreatic cancer, with plasma metabolites serving as mediators. Identifying immune cell phenotypes with potential causal effects on pancreatic cancer sheds light on its underlying mechanisms and suggests novel therapeutic targets.
Collapse
Affiliation(s)
- Ke Zhang
- Dalian Medical University, Dalian, China
| | - Jie Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People’s Hospital Affiliated Yangzhou University, Yangzhou, China
| | - Peng Wang
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People’s Hospital Affiliated Yangzhou University, Yangzhou, China
| | - Yuan Chen
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People’s Hospital Affiliated Yangzhou University, Yangzhou, China
| | - Zhengwang Wang
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People’s Hospital Affiliated Yangzhou University, Yangzhou, China
| | - Xinyu Ge
- Dalian Medical University, Dalian, China
| | - Junqing Wu
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People’s Hospital Affiliated Yangzhou University, Yangzhou, China
| | - Long Chen
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People’s Hospital Affiliated Yangzhou University, Yangzhou, China
| | - Yipin Lu
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People’s Hospital Affiliated Yangzhou University, Yangzhou, China
| | - Peng Xu
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People’s Hospital Affiliated Yangzhou University, Yangzhou, China
| | - Jie Yao
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People’s Hospital Affiliated Yangzhou University, Yangzhou, China
| |
Collapse
|
23
|
van Elsas MJ, Middelburg J, Labrie C, Roelands J, Schaap G, Sluijter M, Tonea R, Ovcinnikovs V, Lloyd K, Schuurman J, Riesenfeld SJ, Gajewski TF, de Miranda NFCC, van Hall T, van der Burg SH. Immunotherapy-activated T cells recruit and skew late-stage activated M1-like macrophages that are critical for therapeutic efficacy. Cancer Cell 2024; 42:1032-1050.e10. [PMID: 38759656 DOI: 10.1016/j.ccell.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 02/23/2024] [Accepted: 04/24/2024] [Indexed: 05/19/2024]
Abstract
Total tumor clearance through immunotherapy is associated with a fully coordinated innate and adaptive immune response, but knowledge on the exact contribution of each immune cell subset is limited. We show that therapy-induced intratumoral CD8+ T cells recruited and skewed late-stage activated M1-like macrophages, which were critical for effective tumor control in two different murine models of cancer immunotherapy. The activated CD8+ T cells summon these macrophages into the tumor and their close vicinity via CCR5 signaling. Exposure of non-polarized macrophages to activated T cell supernatant and tumor lysate recapitulates the late-stage activated and tumoricidal phenotype in vitro. The transcriptomic signature of these macrophages is also detected in a similar macrophage population present in human tumors and coincides with clinical response to immune checkpoint inhibitors. The requirement of a functional co-operation between CD8+ T cells and effector macrophages for effective immunotherapy gives warning to combinations with broad macrophage-targeting strategies.
Collapse
Affiliation(s)
- Marit J van Elsas
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden 2333ZA, the Netherlands
| | - Jim Middelburg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden 2333ZA, the Netherlands
| | - Camilla Labrie
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden 2333ZA, the Netherlands
| | - Jessica Roelands
- Department of Pathology, Leiden University Medical Center, Leiden 2333ZA, the Netherlands
| | - Gaby Schaap
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden 2333ZA, the Netherlands
| | - Marjolein Sluijter
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden 2333ZA, the Netherlands
| | - Ruxandra Tonea
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA; Pritzker School of Molecular Engineering, Chicago, IL 60637, USA
| | | | | | | | | | - Thomas F Gajewski
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Noel F C C de Miranda
- Department of Pathology, Leiden University Medical Center, Leiden 2333ZA, the Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden 2333ZA, the Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden 2333ZA, the Netherlands.
| |
Collapse
|
24
|
Ullah A, Jiao W, Shen B. The role of proinflammatory cytokines and CXC chemokines (CXCL1-CXCL16) in the progression of prostate cancer: insights on their therapeutic management. Cell Mol Biol Lett 2024; 29:73. [PMID: 38745115 PMCID: PMC11094955 DOI: 10.1186/s11658-024-00591-9] [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: 08/30/2023] [Accepted: 05/06/2024] [Indexed: 05/16/2024] Open
Abstract
Reproductive cancers are malignancies that develop in the reproductive organs. One of the leading cancers affecting the male reproductive system on a global scale is prostate cancer (PCa). The negative consequences of PCa metastases endure and are severe, significantly affecting mortality and life quality for those who are affected. The association between inflammation and PCa has captured interest for a while. Inflammatory cells, cytokines, CXC chemokines, signaling pathways, and other elements make up the tumor microenvironment (TME), which is characterized by inflammation. Inflammatory cytokines and CXC chemokines are especially crucial for PCa development and prognosis. Cytokines (interleukins) and CXC chemokines such as IL-1, IL-6, IL-7, IL-17, TGF-β, TNF-α, CXCL1-CXCL6, and CXCL8-CXCL16 are thought to be responsible for the pleiotropic effects of PCa, which include inflammation, progression, angiogenesis, leukocyte infiltration in advanced PCa, and therapeutic resistance. The inflammatory cytokine and CXC chemokines systems are also promising candidates for PCa suppression and immunotherapy. Therefore, the purpose of this work is to provide insight on how the spectra of inflammatory cytokines and CXC chemokines evolve as PCa develops and spreads. We also discussed recent developments in our awareness of the diverse molecular signaling pathways of these circulating cytokines and CXC chemokines, as well as their associated receptors, which may one day serve as PCa-targeted therapies. Moreover, the current status and potential of theranostic PCa therapies based on cytokines, CXC chemokines, and CXC receptors (CXCRs) are examined.
Collapse
Affiliation(s)
- Amin Ullah
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Wang Jiao
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Bairong Shen
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
25
|
Wang ZT, Deng ZM, Dai FF, Yuan MQ, Liu SY, Li BS, Cheng YX. Tumor immunity: A brief overview of tumor‑infiltrating immune cells and research advances into tumor‑infiltrating lymphocytes in gynecological malignancies (Review). Exp Ther Med 2024; 27:166. [PMID: 38476909 PMCID: PMC10928974 DOI: 10.3892/etm.2024.12453] [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: 07/26/2022] [Accepted: 01/03/2023] [Indexed: 03/14/2024] Open
Abstract
Tumor immunity is a promising topic in the area of cancer therapy. The 'soil' function of the tumor microenvironment (TME) for tumor growth has attracted wide attention from scientists. Tumor-infiltrating immune cells in the TME, especially the tumor-infiltrating lymphocytes (TILs), serve a key role in cancer. Firstly, relevant literature was searched in the PubMed and Web of Science databases with the following key words: 'Tumor microenvironment'; 'TME'; 'tumor-infiltrating immunity cells'; 'gynecologic malignancies'; 'the adoptive cell therapy (ACT) of TILs'; and 'TIL-ACT' (https://pubmed.ncbi.nlm.nih.gov/). According to the title and abstract of the articles, relevant items were screened out in the preliminary screening. The most relevant selected items were of two types: All kinds of tumor-infiltrating immune cells; and advanced research on TILs in gynecological malignancies. The results showed that the subsets of TILs were various and complex, while each subpopulation influenced each other and their effects on tumor prognosis were diverse. Moreover, the related research and clinical trials on TILs were mostly concentrated in melanoma and breast cancer, but relatively few focused on gynecological tumors. In conclusion, the present review summarized the biological classification of TILs and the mechanisms of their involvement in the regulation of the immune microenvironment, and subsequently analyzed the development of tumor immunotherapy for TILs. Collectively, the present review provides ideas for the current treatment dilemma of gynecological tumor immune checkpoints, such as adverse reactions, safety, personal specificity and efficacy.
Collapse
Affiliation(s)
- Zi-Tao Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhi-Min Deng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Fang-Fang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Meng-Qin Yuan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Shi-Yi Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Bing-Shu Li
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yan-Xiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
26
|
Bautista E, Jung YH, Jaramillo M, Ganesh H, Varma A, Savsani K, Dakshanamurthy S. AutoPepVax, a Novel Machine-Learning-Based Program for Vaccine Design: Application to a Pan-Cancer Vaccine Targeting EGFR Missense Mutations. Pharmaceuticals (Basel) 2024; 17:419. [PMID: 38675381 PMCID: PMC11053815 DOI: 10.3390/ph17040419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
The current epitope selection methods for peptide vaccines often rely on epitope binding affinity predictions, prompting the need for the development of more sophisticated in silico methods to determine immunologically relevant epitopes. Here, we developed AutoPepVax to expedite and improve the in silico epitope selection for peptide vaccine design. AutoPepVax is a novel program that automatically identifies non-toxic and non-allergenic epitopes capable of inducing tumor-infiltrating lymphocytes by considering various epitope characteristics. AutoPepVax employs random forest classification and linear regression machine-learning-based models, which are trained with datasets derived from tumor samples. AutoPepVax, along with documentation on how to run the program, is freely available on GitHub. We used AutoPepVax to design a pan-cancer peptide vaccine targeting epidermal growth factor receptor (EGFR) missense mutations commonly found in lung adenocarcinoma (LUAD), colorectal adenocarcinoma (CRAD), glioblastoma multiforme (GBM), and head and neck squamous cell carcinoma (HNSCC). These mutations have been previously targeted in clinical trials for EGFR-specific peptide vaccines in GBM and LUAD, and they show promise but lack demonstrated clinical efficacy. Using AutoPepVax, our analysis of 96 EGFR mutations identified 368 potential MHC-I-restricted epitope-HLA pairs from 49,113 candidates and 430 potential MHC-II-restricted pairs from 168,669 candidates. Notably, 19 mutations presented viable epitopes for MHC I and II restrictions. To evaluate the potential impact of a pan-cancer vaccine composed of these epitopes, we used our program, PCOptim, to curate a minimal list of epitopes with optimal population coverage. The world population coverage of our list ranged from 81.8% to 98.5% for MHC Class II and Class I epitopes, respectively. From our list of epitopes, we constructed 3D epitope-MHC models for six MHC-I-restricted and four MHC-II-restricted epitopes, demonstrating their epitope binding potential and interaction with T-cell receptors. AutoPepVax's comprehensive approach to in silico epitope selection addresses vaccine safety, efficacy, and broad applicability. Future studies aim to validate the AutoPepVax-designed vaccines with murine tumor models that harbor the studied mutations.
Collapse
Affiliation(s)
- Enrico Bautista
- Georgetown University School of Medicine, Washington, DC 20007, USA
| | | | | | - Harrish Ganesh
- Virginia Commonwealth University, Richmond, VA 22043, USA
| | - Aryaan Varma
- The George Washington University, Washington, DC 20052, USA
| | - Kush Savsani
- Virginia Commonwealth University, Richmond, VA 22043, USA
| | - Sivanesan Dakshanamurthy
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA
| |
Collapse
|
27
|
Barravecchia I, Lee JM, Manassa J, Magnuson B, Ferris SF, Cavanaugh S, Steele NG, Espinoza CE, Galban CJ, Ramnath N, Frankel TL, Pasca di Magliano M, Galban S. Modeling Molecular Pathogenesis of Idiopathic Pulmonary Fibrosis-Associated Lung Cancer in Mice. Mol Cancer Res 2024; 22:295-307. [PMID: 38015750 PMCID: PMC10906012 DOI: 10.1158/1541-7786.mcr-23-0480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/25/2023] [Accepted: 11/20/2023] [Indexed: 11/30/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by progressive, often fatal loss of lung function due to overactive collagen production and tissue scarring. Patients with IPF have a sevenfold-increased risk of developing lung cancer. The COVID-19 pandemic has increased the number of patients with lung diseases, and infection can worsen prognoses for those with chronic lung diseases and disease-associated cancer. Understanding the molecular pathogenesis of IPF-associated lung cancer is imperative for identifying diagnostic biomarkers and targeted therapies that will facilitate prevention of IPF and progression to lung cancer. To understand how IPF-associated fibroblast activation, matrix remodeling, epithelial-to-mesenchymal transition (EMT), and immune modulation influences lung cancer predisposition, we developed a mouse model to recapitulate the molecular pathogenesis of pulmonary fibrosis-associated lung cancer using the bleomycin and Lewis lung carcinoma models. We demonstrate that development of pulmonary fibrosis-associated lung cancer is likely linked to increased abundance of tumor-associated macrophages and a unique gene signature that supports an immune-suppressive microenvironment through secreted factors. Not surprisingly, preexisting fibrosis provides a pre-metastatic niche and results in augmented tumor growth, and tumors associated with bleomycin-induced fibrosis are characterized by a dramatic loss of cytokeratin expression, indicative of EMT. IMPLICATIONS This characterization of tumors associated with lung diseases provides new therapeutic targets that may aid in the development of treatment paradigms for lung cancer patients with preexisting pulmonary diseases.
Collapse
Affiliation(s)
- Ivana Barravecchia
- Center for Molecular Imaging, The University of Michigan Medical School, Ann Arbor, Michigan
- Department of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan
| | - Jennifer M. Lee
- Center for Molecular Imaging, The University of Michigan Medical School, Ann Arbor, Michigan
- Department of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan
| | - Jason Manassa
- Center for Molecular Imaging, The University of Michigan Medical School, Ann Arbor, Michigan
- Department of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan
| | - Brian Magnuson
- Rogel Cancer Center, The University of Michigan Medical School, Ann Arbor, Michigan
- Department of Biostatistics, School of Public Health, The University of Michigan, Ann Arbor, Michigan
| | - Sarah F. Ferris
- Center for Molecular Imaging, The University of Michigan Medical School, Ann Arbor, Michigan
- Department of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan
| | - Sophia Cavanaugh
- Department of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan
| | - Nina G. Steele
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, Michigan
| | - Carlos E. Espinoza
- Department of Surgery, The University of Michigan Medical School, Ann Arbor, Michigan
| | - Craig J. Galban
- Department of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan
- Department of Biomedical Engineering, The University of Michigan Medical School and College of Engineering, Ann Arbor, Michigan
| | - Nithya Ramnath
- Division of Hematology and Oncology, Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor, Michigan
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Timothy L. Frankel
- Rogel Cancer Center, The University of Michigan Medical School, Ann Arbor, Michigan
- Department of Surgery, The University of Michigan Medical School, Ann Arbor, Michigan
| | - Marina Pasca di Magliano
- Rogel Cancer Center, The University of Michigan Medical School, Ann Arbor, Michigan
- Department of Surgery, The University of Michigan Medical School, Ann Arbor, Michigan
- Department of Cell and Developmental Biology, The University of Michigan Medical School, Ann Arbor, Michigan
| | - Stefanie Galban
- Center for Molecular Imaging, The University of Michigan Medical School, Ann Arbor, Michigan
- Department of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan
- Rogel Cancer Center, The University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
28
|
Hu X, Hu Q, He Y, Yi X, Wu Z, Hu H, Ouyang Y, Yu F, Peng M. Efficacy and safety of microwave ablation and its synergistic potential in the treatment of early-stage non-small cell lung cancer. Clin Imaging 2024; 107:110070. [PMID: 38211397 DOI: 10.1016/j.clinimag.2023.110070] [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: 09/18/2023] [Revised: 12/21/2023] [Accepted: 12/30/2023] [Indexed: 01/13/2024]
Abstract
Lung cancer remains the primary cause of cancer-related mortality globally. In the case of early-stage non-small cell lung cancer (NSCLC), surgical resection, such as lobectomy and sub-lobectomy, continues to be the established standard treatment. However, for patients with insufficient cardiopulmonary function and multiple comorbidities who are unable to undergo surgical resection, nonoperative local therapies, including radiotherapy and thermal ablation, are preferred. In recent years, microwave ablation (MWA) has gained popularity for treating early-stage NSCLC due to its high heating efficiency, good tissue conductance, and heat conduction capabilities. This review provides a comprehensive summary of the current efficacy and safety data regarding MWA for early-stage NSCLC and discusses the potential benefits of combining MWA with other therapies.
Collapse
Affiliation(s)
- Xinhang Hu
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qikang Hu
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yu He
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xuyang Yi
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zeyu Wu
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Huali Hu
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yifan Ouyang
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Fenglei Yu
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Muyun Peng
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
29
|
Bitting RL, Tooze JA, Goodman M, Vile DC, Brown JM, Thomas CY, Neve M, Kooshki M, Addo S, Triozzi PL, Dubey P. Low-dose Paclitaxel with Pembrolizumab Enhances Clinical and Immunologic Responses in Platinum-refractory Urothelial Carcinoma. CANCER RESEARCH COMMUNICATIONS 2024; 4:530-539. [PMID: 38345536 PMCID: PMC10896069 DOI: 10.1158/2767-9764.crc-23-0436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/09/2023] [Accepted: 02/07/2024] [Indexed: 02/28/2024]
Abstract
PURPOSE Single-agent checkpoint inhibition is effective in a minority of patients with platinum-refractory urothelial carcinoma; therefore, the efficacy of combining low-dose paclitaxel with pembrolizumab was tested. MATERIALS AND METHODS This was a prospective, single-arm phase II trial with key inclusion criteria of imaging progression within 12 months of platinum therapy and Eastern Cooperative Oncology Group ≤1. Treatment was pembrolizumab 200 mg day 1 and paclitaxel 80 mg/m2 days 1 and 8 of a 21-day cycle for up to eight cycles unless progression or unacceptable adverse events (AE). The primary endpoint was overall response rate (ORR) with overall survival (OS), 6-month progression-free survival (PFS), and safety as key secondary endpoints. Change in circulating immune cell populations, plasma, and urinary miRs were evaluated. RESULTS Twenty-seven patients were treated between April 2016 and June 2020, with median follow-up of 12.4 months. Baseline median age was 68 years, with 81% men and 78% non-Hispanic White. ORR was 33% by intention to treat and 36% in imaging-evaluable patients with three complete responses. Six-month PFS rate was 48.1% [95% confidence interval (CI): 28.7-65.2] and median OS 12.4 months (95% CI: 8.7 months to not reached). Common ≥ grade 2 possibly-related AEs were anemia, lymphopenia, hyperglycemia, and fatigue; grade 3/4 AEs occurred in 56%, including two immune-mediated AEs (pneumonitis and nephritis). Responding patients had a higher percentage of circulating CD4+IFNγ+ T cells. Levels of some miRs, including plasma miR 181 and miR 223, varied in responders compared with nonresponders. CONCLUSIONS The addition of low-dose paclitaxel to pembrolizumab is active and safe in platinum-refractory urothelial carcinoma. SIGNIFICANCE We found that combining pembrolizumab with low-dose paclitaxel may be effective in patients with urothelial carcinoma progressing on platinum chemotherapy, with favorable safety profiles.
Collapse
Affiliation(s)
- Rhonda L Bitting
- Department of Internal Medicine, Section on Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina
| | - Janet A Tooze
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Michael Goodman
- Department of Internal Medicine, Section on Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina
| | - Donald C Vile
- Department of Internal Medicine, Section on Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jessica M Brown
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio
| | - Christopher Y Thomas
- Department of Internal Medicine, Section on Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina
| | - Morgan Neve
- Department of Internal Medicine, Section on Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Mitra Kooshki
- Department of Internal Medicine, Section on Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Safoa Addo
- Department of Internal Medicine, Section on Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Pierre L Triozzi
- Department of Internal Medicine, Section on Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina
| | - Purnima Dubey
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio
- Pelotonia Institute of Immunooncology, James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
30
|
Bawden EG, Wagner T, Schröder J, Effern M, Hinze D, Newland L, Attrill GH, Lee AR, Engel S, Freestone D, de Lima Moreira M, Gressier E, McBain N, Bachem A, Haque A, Dong R, Ferguson AL, Edwards JJ, Ferguson PM, Scolyer RA, Wilmott JS, Jewell CM, Brooks AG, Gyorki DE, Palendira U, Bedoui S, Waithman J, Hochheiser K, Hölzel M, Gebhardt T. CD4 + T cell immunity against cutaneous melanoma encompasses multifaceted MHC II-dependent responses. Sci Immunol 2024; 9:eadi9517. [PMID: 38241401 DOI: 10.1126/sciimmunol.adi9517] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 12/18/2023] [Indexed: 01/21/2024]
Abstract
Whereas CD4+ T cells conventionally mediate antitumor immunity by providing help to CD8+ T cells, recent clinical studies have implied an important role for cytotoxic CD4+ T cells in cancer immunity. Using an orthotopic melanoma model, we provide a detailed account of antitumoral CD4+ T cell responses and their regulation by major histocompatibility complex class II (MHC II) in the skin. Intravital imaging revealed prominent interactions of CD4+ T cells with tumor debris-laden MHC II+ host antigen-presenting cells that accumulated around tumor cell nests, although direct recognition of MHC II+ melanoma cells alone could also promote CD4+ T cell control. CD4+ T cells stably suppressed or eradicated tumors even in the absence of other lymphocytes by using tumor necrosis factor-α and Fas ligand (FasL) but not perforin-mediated cytotoxicity. Interferon-γ was critical for protection, acting both directly on melanoma cells and via induction of nitric oxide synthase in myeloid cells. Our results illustrate multifaceted and context-specific aspects of MHC II-dependent CD4+ T cell immunity against cutaneous melanoma, emphasizing modulation of this axis as a potential avenue for immunotherapies.
Collapse
Affiliation(s)
- Emma G Bawden
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, Bonn 53105, Germany
| | - Teagan Wagner
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Jan Schröder
- Computational Sciences Initiative, Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Maike Effern
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, Bonn 53105, Germany
| | - Daniel Hinze
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, Bonn 53105, Germany
| | - Lewis Newland
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, Bonn 53105, Germany
| | - Grace H Attrill
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Ariane R Lee
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Sven Engel
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - David Freestone
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Marcela de Lima Moreira
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Elise Gressier
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Nathan McBain
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Annabell Bachem
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Ashraful Haque
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Ruining Dong
- Computational Sciences Initiative, Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
- Department of Clinical Pathology and Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Angela L Ferguson
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- Centenary Institute, University of Sydney, Sydney, NSW, Australia
- Infection, Immunity and Inflammation theme, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Jarem J Edwards
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Peter M Ferguson
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Department of Tissue Oncology and Diagnostic Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
- NSW Health Pathology, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- Department of Tissue Oncology and Diagnostic Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
- NSW Health Pathology, Sydney, NSW, Australia
| | - James S Wilmott
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- United States Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD, USA
- Robert E. Fischell Institute for Biomedical Devices, College Park, MD, USA
- Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, USA
| | - Andrew G Brooks
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - David E Gyorki
- Division of Cancer Surgery, Peter MacCallum Cancer Centre and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre Melbourne, Melbourne, VIC, Australia
| | - Umaimainthan Palendira
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Jason Waithman
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Katharina Hochheiser
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre Melbourne, Melbourne, VIC, Australia
| | - Michael Hölzel
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, Bonn 53105, Germany
| | - Thomas Gebhardt
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
31
|
Régnier P, Vetillard M, Bansard A, Pierre E, Li X, Cagnard N, Gautier EL, Guermonprez P, Manoury B, Podsypanina K, Darrasse-Jèze G. FLT3L-dependent dendritic cells control tumor immunity by modulating Treg and NK cell homeostasis. Cell Rep Med 2023; 4:101256. [PMID: 38118422 PMCID: PMC10772324 DOI: 10.1016/j.xcrm.2023.101256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/05/2023] [Accepted: 10/02/2023] [Indexed: 12/22/2023]
Abstract
FLT3-L-dependent classical dendritic cells (cDCs) recruit anti-tumor and tumor-protecting lymphocytes. We evaluate cancer growth in mice with low, normal, or high levels of cDCs. Paradoxically, both low or high numbers of cDCs improve survival in mice with melanoma. In low cDC context, tumors are restrained by the adaptive immune system through influx of effector T cells and depletion of Tregs and NK cells. High cDC numbers favor the innate anti-tumor response, with massive recruitment of activated NK cells, despite high Treg infiltration. Anti CTLA-4 but not anti PD-1 therapy synergizes with FLT3-L therapy in the cDCHi but not in the cDCLo context. A combination of cDC boost and Treg depletion dramatically improves survival of tumor-bearing mice. Transcriptomic data confirm the paradoxical effect of cDC levels on survival in several human tumor types. cDCHi-TregLo state in such patients predicts best survival. Modulating cDC numbers via FLT3 signaling may have therapeutic potential in human cancer.
Collapse
Affiliation(s)
- Paul Régnier
- Institut Necker Enfants Malades, INSERM U1151, CNRS UMR-8253, Université Paris Cité, Paris, France; Sorbonne Université, INSERM, UMR_S959, Immunology-Immunopathology-Immunotherapy, Paris, France; AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Internal Medicine and Clinical Immunology, DMU3ID, Paris, France
| | - Mathias Vetillard
- Université de Paris Cité, Centre for Inflammation Research, INSERM U1149, CNRS ERL8252, Paris, France; Dendritic Cells and Adaptive Immunity Unit, Institut Pasteur, Paris, France
| | - Adèle Bansard
- Institut Necker Enfants Malades, INSERM U1151, CNRS UMR-8253, Université Paris Cité, Paris, France; Université Paris Cité, Faculté de Médecine, Paris, France
| | | | - Xinyue Li
- Sorbonne Université, INSERM, UMR_S959, Immunology-Immunopathology-Immunotherapy, Paris, France
| | - Nicolas Cagnard
- Structure Fédérative de Recherche Necker, Université Paris Descartes, Paris, France
| | - Emmanuel L Gautier
- Inserm, UMR_S1166, Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France
| | - Pierre Guermonprez
- Université de Paris Cité, Centre for Inflammation Research, INSERM U1149, CNRS ERL8252, Paris, France; Dendritic Cells and Adaptive Immunity Unit, Institut Pasteur, Paris, France
| | - Bénédicte Manoury
- Institut Necker Enfants Malades, INSERM U1151, CNRS UMR-8253, Université Paris Cité, Paris, France
| | - Katrina Podsypanina
- Institut Necker Enfants Malades, INSERM U1151, CNRS UMR-8253, Université Paris Cité, Paris, France; Institut Curie, PSL Research University, CNRS, Sorbonne Université, UMR3664, Paris, France
| | - Guillaume Darrasse-Jèze
- Institut Necker Enfants Malades, INSERM U1151, CNRS UMR-8253, Université Paris Cité, Paris, France; Sorbonne Université, INSERM, UMR_S959, Immunology-Immunopathology-Immunotherapy, Paris, France; Université Paris Cité, Faculté de Médecine, Paris, France.
| |
Collapse
|
32
|
Luo Y, Shreeder B, Jenkins JW, Shi H, Lamichhane P, Zhou K, Bahr DA, Kurian S, Jones KA, Daum JI, Dutta N, Necela BM, Cannon MJ, Block MS, Knutson KL. Th17-inducing dendritic cell vaccines stimulate effective CD4 T cell-dependent antitumor immunity in ovarian cancer that overcomes resistance to immune checkpoint blockade. J Immunother Cancer 2023; 11:e007661. [PMID: 37918918 PMCID: PMC10626769 DOI: 10.1136/jitc-2023-007661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Ovarian cancer (OC), a highly lethal cancer in women, has a 48% 5-year overall survival rate. Prior studies link the presence of IL-17 and Th17 T cells in the tumor microenvironment to improved survival in OC patients. To determine if Th17-inducing vaccines are therapeutically effective in OC, we created a murine model of Th17-inducing dendritic cell (DC) (Th17-DC) vaccination generated by stimulating IL-15 while blocking p38 MAPK in bone marrow-derived DCs, followed by antigen pulsing. METHODS ID8 tumor cells were injected intraperitoneally into mice. Mice were treated with Th17-DC or conventional DC (cDC) vaccine alone or with immune checkpoint blockade (ICB). Systemic immunity, tumor associated immunity, tumor size and survival were examined using a variety of experimental strategies. RESULTS Th17-DC vaccines increased Th17 T cells in the tumor microenvironment, reshaped the myeloid microenvironment, and improved mouse survival compared with cDC vaccines. ICB had limited efficacy in OC, but Th17-inducing DC vaccination sensitized it to anti-PD-1 ICB, resulting in durable progression-free survival by overcoming IL-10-mediated resistance. Th17-DC vaccine efficacy, alone or with ICB, was mediated by CD4 T cells, but not CD8 T cells. CONCLUSIONS These findings emphasize using biologically relevant immune modifiers, like Th17-DC vaccines, in OC treatment to reshape the tumor microenvironment and enhance clinical responses to ICB therapy.
Collapse
Affiliation(s)
- Yan Luo
- Department of Immunology, Mayo Clinic in Florida, Jacksonville, Florida, USA
| | - Barath Shreeder
- Department of Immunology, Mayo Clinic in Florida, Jacksonville, Florida, USA
| | - James W Jenkins
- Department of Immunology, Mayo Clinic in Florida, Jacksonville, Florida, USA
| | - Huashan Shi
- Department of Immunology, Mayo Clinic in Florida, Jacksonville, Florida, USA
| | | | - Kexun Zhou
- Department of Immunology, Mayo Clinic in Florida, Jacksonville, Florida, USA
| | - Deborah A Bahr
- Department of Immunology, Mayo Clinic in Florida, Jacksonville, Florida, USA
| | - Sophia Kurian
- Department of Immunology, Mayo Clinic in Florida, Jacksonville, Florida, USA
| | - Katherine A Jones
- Department of Immunology, Mayo Clinic in Florida, Jacksonville, Florida, USA
| | - Joshua I Daum
- Department of Immunology, Mayo Clinic in Florida, Jacksonville, Florida, USA
| | - Navnita Dutta
- Department of Immunology, Mayo Clinic in Florida, Jacksonville, Florida, USA
| | - Brian M Necela
- Department of Immunology, Mayo Clinic in Florida, Jacksonville, Florida, USA
| | - Martin J Cannon
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Matthew S Block
- Divison of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Keith L Knutson
- Department of Immunology, Mayo Clinic in Florida, Jacksonville, Florida, USA
| |
Collapse
|
33
|
Yang Y, Shi X, Chen G, Qian L. Risk factors for unresectable pancreatic cancer following high-intensity focused ultrasound treatment. Cancer Med 2023; 12:19537-19547. [PMID: 37792639 PMCID: PMC10587952 DOI: 10.1002/cam4.6568] [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: 03/28/2023] [Revised: 08/09/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023] Open
Abstract
PURPOSE Pancreatic cancer is one of the most aggressive malignant tumors with poor prognosis. High-intensity focused ultrasound (HIFU) is an effective and safe treatment option for advanced pancreatic cancer, however, the survival time of patients after the treatment was different. So, the purpose of this study was to evaluate the relationship between the high-risk characteristics and prognosis of unresectable pancreatic cancer after HIFU treatment. PATIENTS AND METHODS This prospective study included 30 patients with unresectable pancreatic cancer who received HIFU at Beijing Friendship Hospital. Data on patients' tumor size, pain scores, peripheral blood lymphocyte subsets, CA19-9 and contrast enhanced ultrasound (CEUS) features were collected to assess the relationship with overall survival (OS) after HIFU. RESULTS The median OS from the start of HIFU treatment was 159 days, 95% confidence interval (95% CI): 108-210. The levels of pain were determined by visual analogue scale (VAS) score, and the quartile of the score decreased from 6 (2, 7) to 4 (2, 5) immediately after one session of the treatment (p = 0.001). The diagnostic model showed that high post VAS score and decreasing of peripheral CD4+ T cells were significantly correlated with poor prognosis (p < 0.05), and showed good discrimination ability (AUC = 0.848, 95% CI = 0.709-0.987). CONCLUSION HIFU can effectively relieve pain in patients with unresectable pancreatic cancer. Post treatment VAS and change of peripheral CD4+ T cells are independent risk factors affecting the prognosis in patients with unresectable pancreatic cancer after HIFU treatment.
Collapse
Affiliation(s)
- Yu Yang
- Department of Ultrasound, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Xian‐quan Shi
- Department of Ultrasound, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Guang Chen
- Department of Interventional Radiology, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Lin‐xue Qian
- Department of Ultrasound, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
34
|
Xie L, Fang J, Yu J, Zhang W, He Z, Ye L, Wang H. The role of CD4 + T cells in tumor and chronic viral immune responses. MedComm (Beijing) 2023; 4:e390. [PMID: 37829505 PMCID: PMC10565399 DOI: 10.1002/mco2.390] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023] Open
Abstract
Immunotherapies are mainly aimed to promote a CD8+ T cell response rather than a CD4+ T cell response as cytotoxic T lymphocytes (CTLs) can directly kill target cells. Recently, CD4+ T cells have received more attention due to their diverse roles in tumors and chronic viral infections. In antitumor and antichronic viral responses, CD4+ T cells relay help signals through dendritic cells to indirectly regulate CD8+ T cell response, interact with B cells or macrophages to indirectly modulate humoral immunity or macrophage polarization, and inhibit tumor blood vessel formation. Additionally, CD4+ T cells can also exhibit direct cytotoxicity toward target cells. However, regulatory T cells exhibit immunosuppression and CD4+ T cells become exhausted, which promote tumor progression and chronic viral persistence. Finally, we also outline immunotherapies based on CD4+ T cells, including adoptive cell transfer, vaccines, and immune checkpoint blockade. Overall, this review summarizes diverse roles of CD4+ T cells in the antitumor or protumor and chronic viral responses, and also highlights the immunotherapies based on CD4+ T cells, giving a better understanding of their roles in tumors and chronic viral infections.
Collapse
Affiliation(s)
- Luoyingzi Xie
- Institute of Hepatopancreatobiliary SurgeryChongqing General HospitalChongqingChina
- The Institute of ImmunologyThird Military Medical University (Army Medical University)ChongqingChina
| | - Jingyi Fang
- The Institute of ImmunologyThird Military Medical University (Army Medical University)ChongqingChina
| | - Juncheng Yu
- Department of Thoracic SurgeryXinqiao Hospital Third Military Medical University (Army Medical University)ChongqingChina
| | - Weinan Zhang
- Department of Plastic & Cosmetic SurgeryArmy Medical Center of PLAAmy Medical UniversityChongqingChina
| | - Zhiqiang He
- Department of Plastic & Cosmetic SurgeryArmy Medical Center of PLAAmy Medical UniversityChongqingChina
| | - Lilin Ye
- The Institute of ImmunologyThird Military Medical University (Army Medical University)ChongqingChina
| | - Huaizhi Wang
- Institute of Hepatopancreatobiliary SurgeryChongqing General HospitalChongqingChina
| |
Collapse
|
35
|
Ramirez F, Zambrano A, Hennis R, Holland N, Lakshmanaswamy R, Chacon J. Sending a Message: Use of mRNA Vaccines to Target the Tumor Immune Microenvironment. Vaccines (Basel) 2023; 11:1465. [PMID: 37766141 PMCID: PMC10534833 DOI: 10.3390/vaccines11091465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/25/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
While cancer immunotherapies have become central to treatment, challenges associated with the ability of tumors to evade the immune system remain significant obstacles. At the heart of this issue is the tumor immune microenvironment, the complex interplay of the tumor microenvironment and the immune response. Recent advances in mRNA cancer vaccines represent major progress towards overcoming some of the challenges posed by deleterious components of the tumor immune microenvironment. Indeed, major breakthroughs in mRNA vaccine technology, such as the use of replacement nucleotides and lipid nanoparticle delivery, led to the vital success of mRNA vaccine technology in fighting COVID-19. This has in turn generated massive additional interest and investment in the platform. In this review, we detail recent research in the nature of the tumor immune microenvironment and in mRNA cancer vaccines and discuss applications by which mRNA cancer vaccines, often in combination with various adjuvants, represent major areas of potential in overcoming tumor immune microenvironment-imposed obstacles. To this end, we also review current mRNA cancer vaccine clinical trials.
Collapse
Affiliation(s)
- Fabiola Ramirez
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.R.); (A.Z.); (R.H.); (N.H.); (R.L.)
| | - Angelica Zambrano
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.R.); (A.Z.); (R.H.); (N.H.); (R.L.)
| | - Robert Hennis
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.R.); (A.Z.); (R.H.); (N.H.); (R.L.)
| | - Nathan Holland
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.R.); (A.Z.); (R.H.); (N.H.); (R.L.)
| | - Rajkumar Lakshmanaswamy
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.R.); (A.Z.); (R.H.); (N.H.); (R.L.)
- L. Frederick Francis Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Jessica Chacon
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.R.); (A.Z.); (R.H.); (N.H.); (R.L.)
| |
Collapse
|
36
|
Li F, Qiu L, Gao Q, Yu L, Liu H, Xue Z, Tao A. Comprehensive bioinformatics analysis combined with experimental validation to screen biomarkers for malignant transformation of oral leukoplakia. Genomics 2023; 115:110686. [PMID: 37454941 DOI: 10.1016/j.ygeno.2023.110686] [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: 03/16/2023] [Revised: 06/26/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
Oral leukoplakia (OLK) is the most common potentially malignant disorders in the oral cavity. This study aimed to screen the key genes of OLK malignant transformation using the Gene Expression Omnibus (GEO) database and experiments. In this study, the GEO database was employed to screen OLK malignant transformation-related genes, which were subsequently identified with a series of bioinformatic analyses. External validation showed that the model based on LAPTM4B, NR3C1, and COX6A1 had high accuracy in diagnosing OLK malignant transformation. Furthermore, the DMBA-induced potentially malignant disorders and OSCC models in vivo and real-time PCR experiment in vitro further verified the database analysis results. In conclusion, three key genes (LAPTM4B, NR3C1, and COX6A1) were screened as potential biomarkers for the diagnosis and treatment of OLK malignant transformation.
Collapse
Affiliation(s)
- Fengji Li
- Department of Stomatology, Shenzhen Luohu Hospital of Traditional Chinese Medicine, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen 518001, China.
| | - Lin Qiu
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, China
| | - Qian Gao
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, China
| | - Liguang Yu
- Lingzhushan Community Health Service Center of Qingdao West Coast New District, Qingdao 266520, China
| | - Han Liu
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing 100051, China
| | - Zhijun Xue
- Department II of Endodontics, Tianjin Stomatological Hospital, Tianjin 300041, China
| | - Anqi Tao
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, China.
| |
Collapse
|
37
|
Bossio SN, Abrate C, Tosello Boari J, Rodriguez C, Canale FP, Ramello MC, Brunotto V, Richer W, Rocha D, Sedlik C, Vincent-Salomon A, Borcoman E, Del Castillo A, Gruppi A, Fernandez E, Acosta Rodríguez EV, Piaggio E, Montes CL. CD39 + conventional CD4 + T cells with exhaustion traits and cytotoxic potential infiltrate tumors and expand upon CTLA-4 blockade. Oncoimmunology 2023; 12:2246319. [PMID: 37885970 PMCID: PMC10599196 DOI: 10.1080/2162402x.2023.2246319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/03/2023] [Accepted: 08/05/2023] [Indexed: 10/28/2023] Open
Abstract
Conventional CD4+ T (Tconv) lymphocytes play important roles in tumor immunity; however, their contribution to tumor elimination remains poorly understood. Here, we describe a subset of tumor-infiltrating Tconv cells characterized by the expression of CD39. In several mouse cancer models, we observed that CD39+ Tconv cells accumulated in tumors but were absent in lymphoid organs. Compared to tumor CD39- counterparts, CD39+ Tconv cells exhibited a cytotoxic and exhausted signature at the transcriptomic level, confirmed by high protein expression of inhibitory receptors and transcription factors related to the exhaustion. Additionally, CD39+ Tconv cells showed increased production of IFNγ , granzyme B, perforin and CD107a expression, but reduced production of TNF. Around 55% of OVA-specific Tconv from B16-OVA tumor-bearing mice, expressed CD39. In vivo CTLA-4 blockade induced the expansion of tumor CD39+ Tconv cells, which maintained their cytotoxic and exhausted features. In breast cancer patients, CD39+ Tconv cells were found in tumors and in metastatic lymph nodes but were less frequent in adjacent non-tumoral mammary tissue and not detected in non-metastatic lymph nodes and blood. Human tumor CD39+ Tconv cells constituted a heterogeneous cell population with features of exhaustion, high expression of inhibitory receptors and CD107a. We found that high CD4 and ENTPD1 (CD39) gene expression in human tumor tissues correlated with a higher overall survival rate in breast cancer patients. Our results identify CD39 as a biomarker of Tconv cells, with characteristics of both exhaustion and cytotoxic potential, and indicate CD39+ Tconv cells as players within the immune response against tumors.
Collapse
Affiliation(s)
- Sabrina N. Bossio
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Córdoba, Argentina
| | - Carolina Abrate
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Córdoba, Argentina
| | - Jimena Tosello Boari
- Institut Curie Research Center, Translational Research Department, INSERM U932, PSL Research University, Paris, France
| | - Constanza Rodriguez
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Córdoba, Argentina
| | - Fernando P. Canale
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Córdoba, Argentina
| | - María C. Ramello
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Córdoba, Argentina
| | - Valentina Brunotto
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Córdoba, Argentina
| | - Wilfrid Richer
- Institut Curie Research Center, Translational Research Department, INSERM U932, PSL Research University, Paris, France
| | - Dario Rocha
- Centro de Investigación y desarrollo en inmunología y enfermedades infecciosas (CIDIE-CONICET), Argentina
| | - Christine Sedlik
- Institut Curie Research Center, Translational Research Department, INSERM U932, PSL Research University, Paris, France
| | - Anne Vincent-Salomon
- Diagnostic and Theranostic Medicine Division, Institut Curie, PSL Research University, Paris, France
| | - Edith Borcoman
- Department of Medical Oncology, Institut Curie, Paris, France
| | | | - Adriana Gruppi
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Córdoba, Argentina
| | - Elmer Fernandez
- Centro de Investigación y desarrollo en inmunología y enfermedades infecciosas (CIDIE-CONICET), Argentina
| | - Eva V. Acosta Rodríguez
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Córdoba, Argentina
| | - Eliane Piaggio
- Institut Curie Research Center, Translational Research Department, INSERM U932, PSL Research University, Paris, France
| | - Carolina L. Montes
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Córdoba, Argentina
| |
Collapse
|
38
|
Buonaguro L, Tagliamonte M. Peptide-based vaccine for cancer therapies. Front Immunol 2023; 14:1210044. [PMID: 37654484 PMCID: PMC10467431 DOI: 10.3389/fimmu.2023.1210044] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/31/2023] [Indexed: 09/02/2023] Open
Abstract
Different strategies based on peptides are available for cancer treatment, in particular to counter-act the progression of tumor growth and disease relapse. In the last decade, in the context of therapeutic strategies against cancer, peptide-based vaccines have been evaluated in different tumor models. The peptides selected for cancer vaccine development can be classified in two main type: tumor-associated antigens (TAAs) and tumor-specific antigens (TSAs), which are captured, internalized, processed and presented by antigen-presenting cells (APCs) to cell-mediated immunity. Peptides loaded onto MHC class I are recognized by a specific TCR of CD8+ T cells, which are activated to exert their cytotoxic activity against tumor cells presenting the same peptide-MHC-I complex. This process is defined as active immunotherapy as the host's immune system is either de novo activated or restimulated to mount an effective, tumor-specific immune reaction that may ultimately lead to tu-mor regression. However, while the preclinical data have frequently shown encouraging results, therapeutic cancer vaccines clinical trials, including those based on peptides have not provided satisfactory data to date. The limited efficacy of peptide-based cancer vaccines is the consequence of several factors, including the identification of specific target tumor antigens, the limited immunogenicity of peptides and the highly immunosuppressive tumor microenvironment (TME). An effective cancer vaccine can be developed only by addressing all such different aspects. The present review describes the state of the art for each of such factors.
Collapse
Affiliation(s)
| | - Maria Tagliamonte
- Innovative Immunological Models Unit, Istituto Nazionale Tumori - IRCCS - “Fond G. Pascale”, Naples, Italy
| |
Collapse
|
39
|
Turner RJ, Guy TV, Geraghty NJ, Splitt A, Watson D, Brungs D, Carolan MG, Miller AA, de Leon JF, Aghmesheh M, Sluyter R. Low Pretreatment CD4 +:CD8 + T Cell Ratios and CD39 +CD73 +CD19 + B Cell Proportions Are Associated with Improved Relapse-Free Survival in Head and Neck Squamous Cell Carcinoma. Int J Mol Sci 2023; 24:12538. [PMID: 37628721 PMCID: PMC10454544 DOI: 10.3390/ijms241612538] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
The ectonucleotidases CD39 and CD73 are present on immune cells and play important roles in cancer progression by suppressing antitumour immunity. As such, CD39 and CD73 on peripheral blood mononuclear cells (PBMCs) are emerging as potential biomarkers to predict disease outcomes and treatment responses in cancer patients. This study aimed to examine T and B cells, including CD39 and CD73 expressing subsets, by flow cytometry in PBMCs from 28 patients with head and neck squamous cell carcinoma (HNSCC) and to assess the correlation with the treatment modality, human papillomavirus (HPV) status, and relapse-free survival (RFS). The PBMCs were examined pre-, mid-, and post-radiotherapy with concurrent cisplatin chemotherapy or anti-epidermal growth factor receptor antibody (cetuximab) therapy. Combination radiotherapy caused changes to T and B cell populations, including CD39 and CD73 expressing subsets, but no such differences were observed between concurrent chemotherapy and cetuximab. Pretreatment PBMCs from HPV+ patients contained increased proportions of CD39-CD73-CD4+ T cells and reduced proportions of CD39-/+CD73+CD4+ T cells compared to the equivalent cells from HPV- patients. Notably, the pretreatment CD4+:CD8+ T cell ratios and CD39+CD73+CD19+ B cell proportions below the respective cohort medians corresponded with an improved RFS. Collectively, this study supports the notion that CD39 and CD73 may contribute to disease outcomes in HNSCC patients and may assist as biomarkers, either alone or as part of immune signatures, in HNSCC. Further studies of CD39 and CD73 on PBMCs from larger cohorts of HNSCC patients are warranted.
Collapse
Affiliation(s)
- Ross J. Turner
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (R.J.T.); (N.J.G.); (D.W.)
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia;
| | - Thomas V. Guy
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia;
| | - Nicholas J. Geraghty
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (R.J.T.); (N.J.G.); (D.W.)
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia;
| | - Ashleigh Splitt
- Illawarra Cancer Care Centre, Wollongong Hospital, Wollongong, NSW 2500, Australia; (A.S.); (D.B.); (M.G.C.); (A.A.M.); (M.A.)
| | - Debbie Watson
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (R.J.T.); (N.J.G.); (D.W.)
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia;
| | - Daniel Brungs
- Illawarra Cancer Care Centre, Wollongong Hospital, Wollongong, NSW 2500, Australia; (A.S.); (D.B.); (M.G.C.); (A.A.M.); (M.A.)
- Graduate School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Martin G. Carolan
- Illawarra Cancer Care Centre, Wollongong Hospital, Wollongong, NSW 2500, Australia; (A.S.); (D.B.); (M.G.C.); (A.A.M.); (M.A.)
| | - Andrew A. Miller
- Illawarra Cancer Care Centre, Wollongong Hospital, Wollongong, NSW 2500, Australia; (A.S.); (D.B.); (M.G.C.); (A.A.M.); (M.A.)
| | | | - Morteza Aghmesheh
- Illawarra Cancer Care Centre, Wollongong Hospital, Wollongong, NSW 2500, Australia; (A.S.); (D.B.); (M.G.C.); (A.A.M.); (M.A.)
| | - Ronald Sluyter
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (R.J.T.); (N.J.G.); (D.W.)
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia;
| |
Collapse
|
40
|
Qayoom H, Sofi S, Mir MA. Targeting tumor microenvironment using tumor-infiltrating lymphocytes as therapeutics against tumorigenesis. Immunol Res 2023; 71:588-599. [PMID: 37004645 DOI: 10.1007/s12026-023-09376-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 03/25/2023] [Indexed: 04/04/2023]
Abstract
The immune system plays a vital role in suppressing tumor cell progression. The tumor microenvironment augmented with significant levels of tumor-infiltrating lymphocytes has been widely investigated and it is suggested that tumor-infiltrating lymphocytes have shown a significant role in the prognosis of cancer patients. Compared to ordinary non-infiltrating lymphocytes, tumor-infiltrating lymphocytes (TILs) are a significant population of lymphocytes that infiltrate tumor tissue and have a higher level of specific immunological reactivity against tumor cells. They serve as an effective immunological defense against various malignancies. TILs are a diverse group of immune cells that are divided into immune subsets based on the pathological and physiological impact they have on the immune system. TILs mainly consist of B-cells, T-cells, or natural killer cells with diverse phenotypic and functional properties. TILs are known to be superior to other immune cells in that they can recognize a wide range of heterogeneous tumor antigens by producing many clones of T cell receptors (TCRs), outperforming treatments like TCR-T cell and CAR-T therapy. With the introduction of genetic engineering technologies, tumor-infiltrating lymphocytes (TILs) have become a ground-breaking therapeutic option for malignancies, but because of the hindrances opposed by the immune microenvironment and the mutation of antigens, the development of TILs as therapeutic has been hindered. By giving some insight into the many variables, such as the various barriers inhibiting its usage as a potential therapeutic agent, we have examined various aspects of TILs in this work.
Collapse
Affiliation(s)
- Hina Qayoom
- Department of Bioresources, School of Biological Sciences, University of Kashmir, 190006, Jammu and Kashmir, India
| | - Shazia Sofi
- Department of Bioresources, School of Biological Sciences, University of Kashmir, 190006, Jammu and Kashmir, India
| | - Manzoor A Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, 190006, Jammu and Kashmir, India.
| |
Collapse
|
41
|
Espinosa-Carrasco G, Scrivo A, Zumbo P, Dave A, Betel D, Hellmann M, Burt BM, Lee HS, Schietinger A. Intratumoral immune triads are required for adoptive T cell therapy-mediated elimination of solid tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.03.547423. [PMID: 37461721 PMCID: PMC10349998 DOI: 10.1101/2023.07.03.547423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Tumor-reactive CD8 T cells found in cancer patients are frequently dysfunctional, unable to halt tumor growth. Adoptive T cell transfer (ACT), the administration of large numbers of in vitro-generated cytolytic tumor-reactive CD8 T cells, is an important cancer immune therapy being pursued. However, a limitation of ACT is that transferred CD8 T cells often rapidly lose effector function, and despite exciting results in certain malignancies, few ACT clinical trials have shown responses in solid tumors. Here, we developed preclinical cancer mouse models to investigate if and how tumor-specific CD4 T cells can be enlisted to overcome CD8 T cell dysfunction in the setting of ACT. In situ confocal microscopy of color-coded cancer cells, tumor-specific CD8 and CD4 T cells, and antigen presenting cells (APC), combined with functional studies, revealed that the spatial positioning and interactions of CD8 and CD4 T cells, but not their numbers, dictates ACT efficacy and anti-tumor responses. We uncover a new role of antigen-specific CD4 T cells in addition to the known requirement for CD4 T cells during priming/activation of naïve CD8 T cells. CD4 T cells must co-engage with CD8 T cells and APC cross-presenting CD8- and CD4-tumor antigens during the effector phase, forming a three-cell-cluster (triad), to license CD8 T cell cytotoxicity and mediate cancer cell elimination. Triad formation transcriptionally and epigenetically reprogram CD8 T cells, prevent T cell dysfunction/exhaustion, and ultimately lead to the elimination of large established tumors and confer long-term protection from recurrence. When intratumoral triad formation was disrupted, adoptively transferred CD8 T cells could not be reprogrammed, and tumors progressed despite equal numbers of tumor-infiltrating CD8 and CD4 T cells. Strikingly, the formation of CD4 T cell::CD8 T cell::APC triads in tumors of patients with lung cancers treated with immune checkpoint blockade was associated with clinical responses, but not CD4::APC dyads or overall numbers of CD8 or CD4 T cells, demonstrating the importance of triads in non-ACT settings in humans. Our work uncovers intratumoral triads as a key requirement for anti-tumor immunity and a new role for CD4 T cells in CD8 T cell cytotoxicity and cancer cell eradication.
Collapse
Affiliation(s)
| | - Aurora Scrivo
- Department of Developmental and Molecular Biology, and Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Paul Zumbo
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Asim Dave
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Doron Betel
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Matthew Hellmann
- Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Bryan M Burt
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX
| | - Hyun-Sung Lee
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX
| | - Andrea Schietinger
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| |
Collapse
|
42
|
Sun L, Su Y, Jiao A, Wang X, Zhang B. T cells in health and disease. Signal Transduct Target Ther 2023; 8:235. [PMID: 37332039 PMCID: PMC10277291 DOI: 10.1038/s41392-023-01471-y] [Citation(s) in RCA: 266] [Impact Index Per Article: 133.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 06/20/2023] Open
Abstract
T cells are crucial for immune functions to maintain health and prevent disease. T cell development occurs in a stepwise process in the thymus and mainly generates CD4+ and CD8+ T cell subsets. Upon antigen stimulation, naïve T cells differentiate into CD4+ helper and CD8+ cytotoxic effector and memory cells, mediating direct killing, diverse immune regulatory function, and long-term protection. In response to acute and chronic infections and tumors, T cells adopt distinct differentiation trajectories and develop into a range of heterogeneous populations with various phenotype, differentiation potential, and functionality under precise and elaborate regulations of transcriptional and epigenetic programs. Abnormal T-cell immunity can initiate and promote the pathogenesis of autoimmune diseases. In this review, we summarize the current understanding of T cell development, CD4+ and CD8+ T cell classification, and differentiation in physiological settings. We further elaborate the heterogeneity, differentiation, functionality, and regulation network of CD4+ and CD8+ T cells in infectious disease, chronic infection and tumor, and autoimmune disease, highlighting the exhausted CD8+ T cell differentiation trajectory, CD4+ T cell helper function, T cell contributions to immunotherapy and autoimmune pathogenesis. We also discuss the development and function of γδ T cells in tissue surveillance, infection, and tumor immunity. Finally, we summarized current T-cell-based immunotherapies in both cancer and autoimmune diseases, with an emphasis on their clinical applications. A better understanding of T cell immunity provides insight into developing novel prophylactic and therapeutic strategies in human diseases.
Collapse
Affiliation(s)
- Lina Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Anjun Jiao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Xin Wang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China.
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China.
| |
Collapse
|
43
|
Huang H, Li Z, Xia Y, Zhao Z, Wang D, Jin H, Liu F, Yang Y, Shen L, Lu Z. Association between radiomics features of DCE-MRI and CD8 + and CD4 + TILs in advanced gastric cancer. Pathol Oncol Res 2023; 29:1611001. [PMID: 37342362 PMCID: PMC10277864 DOI: 10.3389/pore.2023.1611001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 05/24/2023] [Indexed: 06/22/2023]
Abstract
Objective: The aim of this investigation was to explore the correlation between the levels of tumor-infiltrating CD8+ and CD4+ T cells and the quantitative pharmacokinetic parameters of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) in patients with advanced gastric cancer. Methods: We retrospectively analyzed the data of 103 patients with histopathologically confirmed advanced gastric cancer (AGC). Three pharmacokinetic parameters, Kep, Ktrans, and Ve, and their radiomics characteristics were obtained by Omni Kinetics software. Immunohistochemical staining was used to determine CD4+ and CD8+ TILs. Statistical analysis was subsequently performed to assess the correlation between radiomics characteristics and CD4+ and CD8+ TIL density. Results: All patients included in this study were finally divided into either a CD8+ TILs low-density group (n = 51) (CD8+ TILs < 138) or a high-density group (n = 52) (CD8+ TILs ≥ 138), and a CD4+ TILs low-density group (n = 51) (CD4+ TILs < 87) or a high-density group (n = 52) (CD4+ TILs ≥ 87). ClusterShade and Skewness based on Kep and Skewness based on Ktrans both showed moderate negative correlation with CD8+ TIL levels (r = 0.630-0.349, p < 0.001), with ClusterShade based on Kep having the highest negative correlation (r = -0.630, p < 0.001). Inertia-based Kep showed a moderate positive correlation with the CD4+ TIL level (r = 0.549, p < 0.001), and the Correlation based on Kep showed a moderate negative correlation with the CD4+ TIL level, which also had the highest correlation coefficient (r = -0.616, p < 0.001). The diagnostic efficacy of the above features was assessed by ROC curves. For CD8+ TILs, ClusterShade of Kep had the highest mean area under the curve (AUC) (0.863). For CD4+ TILs, the Correlation of Kep had the highest mean AUC (0.856). Conclusion: The radiomics features of DCE-MRI are associated with the expression of tumor-infiltrating CD8+ and CD4+ T cells in AGC, which have the potential to noninvasively evaluate the expression of CD8+ and CD4+ TILs in AGC patients.
Collapse
Affiliation(s)
- Huizhen Huang
- Shaoxing of Medicine, Shaoxing University, Shaoxing, China
| | - Zhiheng Li
- Department of Radiology, Anhui Provincial Hospital, Hefei, China
| | - Yue Xia
- Shaoxing of Medicine, Shaoxing University, Shaoxing, China
| | - Zhenhua Zhao
- Department of Radiology, Shaoxing People’s Hospital, Shaoxing, China
| | - Dandan Wang
- Department of Radiology, Shaoxing People’s Hospital, Shaoxing, China
| | - Hongyan Jin
- Country Department of Pathology, Shaoxing People’s Hospital, Shaoxing, China
| | - Fang Liu
- Country Department of Pathology, Shaoxing People’s Hospital, Shaoxing, China
| | - Ye Yang
- Country Department of Pathology, Shaoxing People’s Hospital, Shaoxing, China
| | - Liyijing Shen
- Department of Radiology, Shaoxing People’s Hospital, Shaoxing, China
| | - Zengxin Lu
- Department of Radiology, Shaoxing People’s Hospital, Shaoxing, China
- The First Affiliated Hospital of Shaoxing University, Shaoxing, China
| |
Collapse
|
44
|
Drougkas K, Karampinos K, Karavolias I, Koumprentziotis IA, Ploumaki I, Triantafyllou E, Trontzas I, Kotteas E. Comprehensive clinical evaluation of CAR-T cell immunotherapy for solid tumors: a path moving forward or a dead end? J Cancer Res Clin Oncol 2023; 149:2709-2734. [PMID: 36564524 PMCID: PMC10129996 DOI: 10.1007/s00432-022-04547-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/16/2022] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Chimeric Antigen Receptor (CAR)-T cell therapy is a form of adoptive cell therapy that has demonstrated tremendous results in the treatment of hematopoietic malignancies, leading to the US Food and Drug Administration (FDA) approval of four CD19-targeted CAR-T cell products. With the unprecedented success of CAR-T cell therapy in hematological malignancies, hundreds of preclinical studies and clinical trials are currently undergoing to explore the translation of this treatment to solid tumors. However, the clinical experience in non-hematologic malignancies has been less encouraging, with only a few patients achieving complete responses. Tumor-associated antigen heterogeneity, inefficient CAR-T cell trafficking and the immunosuppressive tumor microenvironment are considered as the most pivotal roadblocks in solid tumor CAR-T cell therapy. MATERIALS AND METHODS We reviewed the relevant literature/clinical trials for CAR-T cell immunotherapy for solid tumors from Pubmed and ClinicalTrials.gov. CONCLUSION Herein, we provide an update on solid tumor CAR-T cell clinical trials, focusing on the studies with published results. We further discuss some of the key hurdles that CAR-T cell therapy is encountering for solid tumor treatment as well as the strategies that are exploited to overcome these obstacles.
Collapse
Affiliation(s)
- Konstantinos Drougkas
- Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, 152 Mesogeion Avenue, 11527, Athens, Greece.
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece.
| | - Konstantinos Karampinos
- Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, 152 Mesogeion Avenue, 11527, Athens, Greece
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Ioannis Karavolias
- Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, 152 Mesogeion Avenue, 11527, Athens, Greece
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Ioannis-Alexios Koumprentziotis
- Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, 152 Mesogeion Avenue, 11527, Athens, Greece
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Ioanna Ploumaki
- Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, 152 Mesogeion Avenue, 11527, Athens, Greece
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Efthymios Triantafyllou
- Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, 152 Mesogeion Avenue, 11527, Athens, Greece
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Ioannis Trontzas
- Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, 152 Mesogeion Avenue, 11527, Athens, Greece
- Department of Pathology, Yale University School of Medicine, New Haven, USA, CT
| | - Elias Kotteas
- Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, 152 Mesogeion Avenue, 11527, Athens, Greece
| |
Collapse
|
45
|
Kruse B, Buzzai AC, Shridhar N, Braun AD, Gellert S, Knauth K, Pozniak J, Peters J, Dittmann P, Mengoni M, van der Sluis TC, Höhn S, Antoranz A, Krone A, Fu Y, Yu D, Essand M, Geffers R, Mougiakakos D, Kahlfuß S, Kashkar H, Gaffal E, Bosisio FM, Bechter O, Rambow F, Marine JC, Kastenmüller W, Müller AJ, Tüting T. CD4 + T cell-induced inflammatory cell death controls immune-evasive tumours. Nature 2023; 618:1033-1040. [PMID: 37316667 PMCID: PMC10307640 DOI: 10.1038/s41586-023-06199-x] [Citation(s) in RCA: 127] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 05/11/2023] [Indexed: 06/16/2023]
Abstract
Most clinically applied cancer immunotherapies rely on the ability of CD8+ cytolytic T cells to directly recognize and kill tumour cells1-3. These strategies are limited by the emergence of major histocompatibility complex (MHC)-deficient tumour cells and the formation of an immunosuppressive tumour microenvironment4-6. The ability of CD4+ effector cells to contribute to antitumour immunity independently of CD8+ T cells is increasingly recognized, but strategies to unleash their full potential remain to be identified7-10. Here, we describe a mechanism whereby a small number of CD4+ T cells is sufficient to eradicate MHC-deficient tumours that escape direct CD8+ T cell targeting. The CD4+ effector T cells preferentially cluster at tumour invasive margins where they interact with MHC-II+CD11c+ antigen-presenting cells. We show that T helper type 1 cell-directed CD4+ T cells and innate immune stimulation reprogramme the tumour-associated myeloid cell network towards interferon-activated antigen-presenting and iNOS-expressing tumouricidal effector phenotypes. Together, CD4+ T cells and tumouricidal myeloid cells orchestrate the induction of remote inflammatory cell death that indirectly eradicates interferon-unresponsive and MHC-deficient tumours. These results warrant the clinical exploitation of this ability of CD4+ T cells and innate immune stimulators in a strategy to complement the direct cytolytic activity of CD8+ T cells and natural killer cells and advance cancer immunotherapies.
Collapse
Affiliation(s)
- Bastian Kruse
- Laboratory of Experimental Dermatology, Department of Dermatology, University Hospital and Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke University, Magdeburg, Germany
| | - Anthony C Buzzai
- Laboratory of Experimental Dermatology, Department of Dermatology, University Hospital and Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke University, Magdeburg, Germany
| | - Naveen Shridhar
- Laboratory of Experimental Dermatology, Department of Dermatology, University Hospital and Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke University, Magdeburg, Germany
| | - Andreas D Braun
- Laboratory of Experimental Dermatology, Department of Dermatology, University Hospital and Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke University, Magdeburg, Germany
| | - Susan Gellert
- Laboratory of Experimental Dermatology, Department of Dermatology, University Hospital and Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke University, Magdeburg, Germany
| | - Kristin Knauth
- Laboratory of Experimental Dermatology, Department of Dermatology, University Hospital and Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke University, Magdeburg, Germany
| | - Joanna Pozniak
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Johannes Peters
- Laboratory of Experimental Dermatology, Department of Dermatology, University Hospital and Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke University, Magdeburg, Germany
| | - Paulina Dittmann
- Laboratory of Experimental Dermatology, Department of Dermatology, University Hospital and Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke University, Magdeburg, Germany
| | - Miriam Mengoni
- Laboratory of Experimental Dermatology, Department of Dermatology, University Hospital and Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke University, Magdeburg, Germany
| | - Tetje Cornelia van der Sluis
- Laboratory of Experimental Dermatology, Department of Dermatology, University Hospital and Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke University, Magdeburg, Germany
| | - Simon Höhn
- Laboratory of Experimental Dermatology, Department of Dermatology, University Hospital and Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke University, Magdeburg, Germany
| | - Asier Antoranz
- Translational Cell and Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Anna Krone
- Institute of Molecular and Clinical Immunology, Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke University, Magdeburg, Germany
| | - Yan Fu
- Institute of Molecular and Clinical Immunology, Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke University, Magdeburg, Germany
| | - Di Yu
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Magnus Essand
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Robert Geffers
- Helmholtz Centre for Infection Research, Brunswick, Germany
| | - Dimitrios Mougiakakos
- Department of Hematology, University Hospital and Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke University, Magdeburg, Germany
| | - Sascha Kahlfuß
- Institute of Molecular and Clinical Immunology, Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke University, Magdeburg, Germany
| | - Hamid Kashkar
- Institute for Molecular Immunology, Centre for Molecular Medicine Cologne and Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases, University of Cologne, Cologne, Germany
| | - Evelyn Gaffal
- Laboratory of Experimental Dermatology, Department of Dermatology, University Hospital and Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke University, Magdeburg, Germany
| | | | - Oliver Bechter
- Department of General Medical Oncology, UZ Leuven, Leuven, Belgium
| | - Florian Rambow
- Department of Applied Computational Cancer Research, Institute for AI in Medicine (IKIM), University Hospital Essen, Essen, Germany
- University of Duisburg-Essen, Essen, Germany
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - Andreas J Müller
- Institute of Molecular and Clinical Immunology, Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke University, Magdeburg, Germany.
| | - Thomas Tüting
- Laboratory of Experimental Dermatology, Department of Dermatology, University Hospital and Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
46
|
Xie L, Meng Z. Immunomodulatory effect of locoregional therapy in the tumor microenvironment. Mol Ther 2023; 31:951-969. [PMID: 36694462 PMCID: PMC10124087 DOI: 10.1016/j.ymthe.2023.01.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/15/2022] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Cancer immunotherapy appears to be a promising treatment option; however, only a subset of patients with cancer responds favorably to treatment. Locoregional therapy initiates a local antitumor immune response by disrupting immunosuppressive components, releasing immunostimulatory damage-associated molecular patterns, recruiting immune effectors, and remodeling the tumor microenvironment. Many studies have shown that locoregional therapy can produce specific antitumor immunity alone; nevertheless, the effect is relatively weak and transient. Furthermore, increasing research efforts have explored the potential synergy between locoregional therapy and immunotherapy to enhance the long-term systemic antitumor immune effect and improve survival. Therefore, further research is needed into the immunomodulatory effects of locoregional therapy and immunotherapy to augment antitumor effects. This review article summarizes the key components of the tumor microenvironment, discusses the immunomodulatory role of locoregional therapy in the tumor microenvironment, and emphasizes the therapeutic potential of locoregional therapy in combination with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Lin Xie
- Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Zhiqiang Meng
- Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China.
| |
Collapse
|
47
|
Brown MC, Beasley GM, McKay ZP, Yang Y, Desjardins A, Randazzo DM, Landi D, Ashley DM, Bigner DD, Nair SK, Gromeier M. Intratumor childhood vaccine-specific CD4 + T-cell recall coordinates antitumor CD8 + T cells and eosinophils. J Immunother Cancer 2023; 11:jitc-2022-006463. [PMID: 37072349 PMCID: PMC10124325 DOI: 10.1136/jitc-2022-006463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Antitumor mechanisms of CD4+ T cells remain crudely defined, and means to effectively harness CD4+ T-cell help for cancer immunotherapy are lacking. Pre-existing memory CD4+ T cells hold potential to be leveraged for this purpose. Moreover, the role of pre-existing immunity in virotherapy, particularly recombinant poliovirus immunotherapy where childhood polio vaccine specific immunity is ubiquitous, remains unclear. Here we tested the hypothesis that childhood vaccine-specific memory T cells mediate antitumor immunotherapy and contribute to the antitumor efficacy of polio virotherapy. METHODS The impact of polio immunization on polio virotherapy, and the antitumor effects of polio and tetanus recall were tested in syngeneic murine melanoma and breast cancer models. CD8+ T-cell and B-cell knockout, CD4+ T-cell depletion, CD4+ T-cell adoptive transfer, CD40L blockade, assessments of antitumor T-cell immunity, and eosinophil depletion defined antitumor mechanisms of recall antigens. Pan-cancer transcriptome data sets and polio virotherapy clinical trial correlates were used to assess the relevance of these findings in humans. RESULTS Prior vaccination against poliovirus substantially bolstered the antitumor efficacy of polio virotherapy in mice, and intratumor recall of poliovirus or tetanus immunity delayed tumor growth. Intratumor recall antigens augmented antitumor T-cell function, caused marked tumor infiltration of type 2 innate lymphoid cells and eosinophils, and decreased proportions of regulatory T cells (Tregs). Antitumor effects of recall antigens were mediated by CD4+ T cells, limited by B cells, independent of CD40L, and dependent on eosinophils and CD8+ T cells. An inverse relationship between eosinophil and Treg signatures was observed across The Cancer Genome Atlas (TCGA) cancer types, and eosinophil depletion prevented Treg reductions after polio recall. Pretreatment polio neutralizing antibody titers were higher in patients living longer, and eosinophil levels increased in the majority of patients, after polio virotherapy. CONCLUSION Pre-existing anti-polio immunity contributes to the antitumor efficacy of polio virotherapy. This work defines cancer immunotherapy potential of childhood vaccines, reveals their utility to engage CD4+ T-cell help for antitumor CD8+ T cells, and implicates eosinophils as antitumor effectors of CD4+ T cells.
Collapse
Affiliation(s)
- Michael C Brown
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Georgia M Beasley
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Zachary P McKay
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Yuanfan Yang
- Department of Neurosurgery, University of Alabama Division of Neurosurgery, Birmingham, Alabama, USA
| | - Annick Desjardins
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Dina M Randazzo
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Daniel Landi
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - David M Ashley
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Darell D Bigner
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Smita K Nair
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Matthias Gromeier
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
48
|
Zou Y, Chen Z, Chen Z, Zhang Y, Zheng Z, Wu F, Xu L, Li Q, Lin Y, Shi N, Jin H. Peripheral Th and NK cells are associated with response to lenvatinib plus PD-1 inhibitor in unresectable hepatocellular carcinoma. Clin Immunol 2023; 249:109290. [PMID: 36931486 DOI: 10.1016/j.clim.2023.109290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/18/2023] [Accepted: 03/08/2023] [Indexed: 03/17/2023]
Abstract
The value of peripheral blood lymphocyte subpopulations in predicting responses to lenvatinib combination with programmed death-1 (PD-1) inhibitors in unresectable hepatocellular carcinoma (HCC) was investigated. Fifteen patients received objective responses (OR) and sixteen patients had non-objective responses (NOR) were analyzed. The counts of peripheral blood lymphocyte subpopulations from patients were measured before treatment, second (at week 3), and third doses (at week 6) of the PD-1 inhibitor administration, and correlated with responses. Helper T (Th) cells and natural killers (NK) cells were more abundant in the OR group and found to be important predictors of OR in a stepwise multivariate logistic regression analysis. These cutoff values of Th and NK cells could help to distinguish OR from NOR cases accurately and provide clinical benefits.
Collapse
Affiliation(s)
- Yiping Zou
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zhihong Chen
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Shantou University Medical College, Shantou, China
| | - Zhenrong Chen
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yuanpeng Zhang
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zehao Zheng
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Shantou University Medical College, Shantou, China
| | - Fan Wu
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Lianqun Xu
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qiao Li
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ye Lin
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ning Shi
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Haosheng Jin
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
49
|
Rajendran M, Rao M, Elhence PA, Bharti JN, Singh P, Yadav G, Nalwa A, Goyal AD. Expression of Programmed Cell Death Ligand-1 and Mismatch Repair Status in Endometrial Carcinomas. J Midlife Health 2023; 14:81-86. [PMID: 38029026 PMCID: PMC10664048 DOI: 10.4103/jmh.jmh_6_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/01/2023] [Accepted: 03/07/2023] [Indexed: 12/01/2023] Open
Abstract
Background and Aims Programmed death ligand-1 (PD-L1) is a co-regulatory molecule that suppresses local immunity, and mismatch repair (MMR) deficiency (dMMR) is reported to influence the response to anti-PD-L1-targeted therapy. This study was conducted to find the PD-L1 status, the occurrence of dMMR in endometrial carcinomas, and the association between them. Materials and Methods The study included 35 resected specimens of endometrial carcinomas represented on formalin-fixed paraffin-embedded sections from January 2016 to July 2020. The clinicopathologic information including patient age, tumor histologic type, grade, stage, lymphovascular invasion, the extent of myometrial invasion, and the percentage of tumor-infiltrating lymphocytes (TILs) were obtained in all cases. The expression of PD-L1 and MMR antibodies including mutS homolog 2 (MSH-2), MSH-6, mutL homolog 1 (MLH-1) and MLH-3, and postmeiotic segregation 2 were assessed using immunohistochemistry. The statistical analysis was done using the Statistical Package for the Social Sciences (SPSS) version 26. Results PD-L1 expression was noted in 48.6% of the cases in tumor cells and 65.7% of the cases in TILs and MMR was deficient in 28.6% of endometrial carcinomas. A statistically significant relation was noted between dMMR and TILs, PD-L1 expression in tumor cells and TILs, PD-L1 expression in tumor cells, and extent of myometrial invasion. Although there was no statistically significant association between MMR status and PD-L1 expression in tumor cells or TILs, 60% of patients with dMMR were PD-L1 positive. Conclusion Sixty percent of dMMR cases showed PD-L1 expression in tumor cells. We conclude, ECs that are MMR deficient might get better response to anti-PD-L1 therapy. This study also revealed the prognostic use of TILs in PD-L1-expressed tumors.
Collapse
Affiliation(s)
- Madhubala Rajendran
- Department of Pathology, Aarupadai Veedu Medical College and Hospital, Puducherry, India
| | - Meenakshi Rao
- Department of Pathology and Lab Medicine, AIIMS, Jodhpur, Rajasthan, India
| | | | | | - Pratibha Singh
- Department of Obstetrics and Gynaecology, AIIMS, Jodhpur, Rajasthan, India
| | - Garima Yadav
- Department of Obstetrics and Gynaecology, AIIMS, Jodhpur, Rajasthan, India
| | - Aasma Nalwa
- Department of Pathology and Lab Medicine, AIIMS, Jodhpur, Rajasthan, India
| | - Akhil Dhanesh Goyal
- Department of Community Medicine and Family Medicine, AIIMS, Jodhpur, Rajasthan, India
| |
Collapse
|
50
|
Ahmed H, Mahmud AR, Siddiquee MFR, Shahriar A, Biswas P, Shimul MEK, Ahmed SZ, Ema TI, Rahman N, Khan MA, Mizan MFR, Emran TB. Role of T cells in cancer immunotherapy: Opportunities and challenges. CANCER PATHOGENESIS AND THERAPY 2023; 1:116-126. [PMID: 38328405 PMCID: PMC10846312 DOI: 10.1016/j.cpt.2022.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/11/2022] [Accepted: 12/16/2022] [Indexed: 09/01/2023]
Abstract
Immunotherapies boosting the immune system's ability to target cancer cells are promising for the treatment of various tumor types, yet clinical responses differ among patients and cancers. Recently, there has been increasing interest in novel cancer immunotherapy practices aimed at triggering T cell-mediated anti-tumor responses. Antigen-directed cytotoxicity mediated by T lymphocytes has become a central focal point in the battle against cancer utilizing the immune system. The molecular and cellular mechanisms involved in the actions of T lymphocytes have directed new therapeutic approaches in cancer immunotherapy, including checkpoint blockade, adoptive and chimeric antigen receptor (CAR) T cell therapy, and cancer vaccinology. This review addresses all the strategies targeting tumor pathogenesis, including metabolic pathways, to evaluate the clinical significance of current and future immunotherapies for patients with cancer, which are further engaged in T cell activation, differentiation, and response against tumors.
Collapse
Affiliation(s)
- Hossain Ahmed
- Department of Biotechnology and Genetic Engineering, University of Development Alternative (UODA), 4/4B, Block A, Lalmatia, Dhaka, 1209, Bangladesh
| | - Aar Rafi Mahmud
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail, 1902, Bangladesh
| | | | - Asif Shahriar
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
| | - Partha Biswas
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology (JUST), Jashore, 7408, Bangladesh
| | - Md. Ebrahim Khalil Shimul
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology (JUST), Jashore, 7408, Bangladesh
| | - Shahlaa Zernaz Ahmed
- Department of Biochemistry and Microbiology, North South University, Dhaka, 1229, Bangladesh
| | - Tanzila Ismail Ema
- Department of Biochemistry and Microbiology, North South University, Dhaka, 1229, Bangladesh
| | - Nova Rahman
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - Md. Arif Khan
- Department of Biotechnology and Genetic Engineering, University of Development Alternative (UODA), 4/4B, Block A, Lalmatia, Dhaka, 1209, Bangladesh
| | | | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| |
Collapse
|