1
|
Bauer M, Santos P, Wilfer A, van den Berg E, Zietsman A, Vetter M, Kaufhold S, Wickenhauser C, Dos-Santos-Silva I, Chen WC, Cubasch H, Murugan N, McCormack V, Joffe M, Seliger B, Kantelhardt E. HIV status alters immune cell infiltration and activation profile in women with breast cancer. Nat Commun 2025; 16:4699. [PMID: 40393975 DOI: 10.1038/s41467-025-59408-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 04/23/2025] [Indexed: 05/22/2025] Open
Abstract
The breast cancer (BC)-related mortality is higher and the immunity is altered in women living with HIV (WLWH) compared to HIV-negative women. Therefore, tumor samples of 296 black BC patients from South Africa and Namibia with known age, HIV status, tumor stage, hormone receptor and HER2 status and overall survival (OS) are analyzed for components of the tumor microenvironment (TME). WLWH (n = 117), either with suppressed viral activity (HR = 1.25) or with immune suppression (HR = 2.04), have a shorter OS. HIV status is associated with increased numbers of CD8+ T cells in the TME compared to HIV-negative patients; no correlation is found with CD4+ T cell numbers in the blood. Moreover, an increased expression of CD276/B7-H3 and a more pronounced IFN-γ signaling in the tumors are found in WLWH, independent of age, stage, and BC subtypes. In conclusion, altered T cell composition and CD276 expression in WLWH may contribute to inferior survival and can be used for targeted treatment.
Collapse
Affiliation(s)
- Marcus Bauer
- Institute of Pathology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
- Global and Planetary Health Working Group, Institute of Medical Epidemiology, Biometrics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
| | - Pablo Santos
- Global and Planetary Health Working Group, Institute of Medical Epidemiology, Biometrics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Andreas Wilfer
- Institute of Pathology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Krukenberg Cancer Center, University Hospital Halle, Halle (Saale), Germany
| | - Eunice van den Berg
- Department of Anatomical Pathology, University of the Witwatersrand, National Health Laboratory Service, Johannesburg, South Africa
| | - Annelle Zietsman
- AB May Cancer Centre, Windhoek Central Hospital, Windhoek, Namibia
| | - Martina Vetter
- Department of Gynecology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Sandy Kaufhold
- Department of Gynecology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Claudia Wickenhauser
- Institute of Pathology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Isabel Dos-Santos-Silva
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine (LSHTM), London, UK
| | - Wenlong Carl Chen
- Global and Planetary Health Working Group, Institute of Medical Epidemiology, Biometrics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Strengthening Oncology Services Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Cancer Registry, National Health Laboratory Service, Johannesburg, South Africa
| | - Herbert Cubasch
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nivashini Murugan
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Valerie McCormack
- International Agency for Research on Cancer (IARC/WHO), Environment and Lifestyle Epidemiology Branch, Lyon, France
| | - Maureen Joffe
- Global and Planetary Health Working Group, Institute of Medical Epidemiology, Biometrics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Strengthening Oncology Services Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Noncommunicable Diseases Research Division, Wits Health Consortium (PTY) Ltd, University Witwatersrand, Johannesburg, South Africa
- Strengthening Oncology Services Research Unit,Faculty of Health Sciences, University Witwatersrand, Johannesburg, South Africa
| | - Barbara Seliger
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany.
- Institute of Translational Immunology, Medical School Theodor Fontane, Brandenburg an der Havel, Germany.
| | - Eva Kantelhardt
- Global and Planetary Health Working Group, Institute of Medical Epidemiology, Biometrics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Department of Gynecology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
2
|
Kunika MD, Kannan A, Velasco GJ, Feng Y, Seaman S, Das BK, Pham D, Lambrecht N, Zhao H, St. Croix B, Gao L. m276-SL-PBD eradicates tumors and instigates long-lasting tumor-free survival in Merkel cell carcinoma preclinical models. iScience 2025; 28:112436. [PMID: 40384933 PMCID: PMC12084002 DOI: 10.1016/j.isci.2025.112436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 12/30/2024] [Accepted: 04/10/2025] [Indexed: 05/20/2025] Open
Abstract
Merkel cell carcinoma (MCC) is a rare but aggressive neuroendocrine carcinoma, and immune checkpoint inhibitors (ICIs) are the only approved therapy; nonetheless, resistance is notable and there is a critical need for novel effective therapies. Recently, CD276 was identified as a promising therapeutic target in human cancers. In preclinical studies, a modified CD276 antibody-drug conjugate (ADC) with pyrrolobenzodiazepine (m276-SL-PBD) elicited more potent anti-tumor effects than two CD276 ADCs currently in clinical trials. Here, we uncover notable CD276 expression in MCC patient tumors, and demonstrate m276-SL-PBD efficacy against MCC preclinical models. Complete eradication is observed in all xenografts bearing CD276 expression, with 82% achieving 180-day tumor-free survival after 4 or 5 weekly doses, and m276-SL-PBD remained efficacious against relapsed tumors. Of clinical relevance, m276-SL-PBD retains its potency in MCC-bearing humanized mice. Importantly, no detectable adverse effects were observed. Thus, m276-SL-PBD is a promising therapy for patients unsuitable or resistant to ICIs.
Collapse
Affiliation(s)
- Mikaela D. Kunika
- Southern California Institute for Research and Education, Long Beach, CA 90822, USA
| | - Aarthi Kannan
- Southern California Institute for Research and Education, Long Beach, CA 90822, USA
- Department of Dermatology, University of California-Irvine, Irvine, CA 92697, USA
| | - Graham J. Velasco
- Pathology Department, Tibor Rubin VA Medical Center, VA Long Beach Healthcare System, Long Beach, CA 90822, USA
| | - Yang Feng
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702, USA
| | - Steven Seaman
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702, USA
| | - Bhaba K. Das
- Southern California Institute for Research and Education, Long Beach, CA 90822, USA
| | - Dillon Pham
- Southern California Institute for Research and Education, Long Beach, CA 90822, USA
| | - Nils Lambrecht
- Pathology Department, Tibor Rubin VA Medical Center, VA Long Beach Healthcare System, Long Beach, CA 90822, USA
| | - Haibo Zhao
- Southern California Institute for Research and Education, Long Beach, CA 90822, USA
| | - Brad St. Croix
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702, USA
| | - Ling Gao
- Southern California Institute for Research and Education, Long Beach, CA 90822, USA
- Department of Dermatology, University of California-Irvine, Irvine, CA 92697, USA
- Dermatology Section, Tibor Rubin VA Medical Center, VA Long Beach Healthcare System, Long Beach, CA 90822, USA
| |
Collapse
|
3
|
Bertok T, Pinkeova A, Lorencova L, Datkova A, Hires M, Jane E, Tkac J. Glycoproteomics of Gastrointestinal Cancers and Its Use in Clinical Diagnostics. J Proteome Res 2025. [PMID: 40368336 DOI: 10.1021/acs.jproteome.5c00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Cancer is a leading cause of death worldwide, resulting in substantial economic costs. Because cancer is a complex, heterogeneous group of diseases affecting a variety of cells, its detection may sometimes be difficult. Herein we review a large group of the gastrointestinal cancers (oral, esophageal, stomach, pancreatic, liver, and bowel cancers) and the possibility of using glycans conjugated to protein backbones for less-invasive diagnoses than the commonly used endoscopic approaches. The reality of bacterial N-glycosylation and the effect of epithelial mucosa on gut microbiota are discussed. Current advantages, barriers, and advantages in the prospective use of selected glycomic approaches in clinical practice are also detailed.
Collapse
Affiliation(s)
- Tomas Bertok
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovak Republic
| | - Andrea Pinkeova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovak Republic
- Glycanostics, Kudlakova 7, 841 01 Bratislava, Slovak Republic
| | - Lenka Lorencova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovak Republic
| | - Anna Datkova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovak Republic
| | - Michal Hires
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovak Republic
| | - Eduard Jane
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovak Republic
- Glycanostics, Kudlakova 7, 841 01 Bratislava, Slovak Republic
| | - Jan Tkac
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovak Republic
- Glycanostics, Kudlakova 7, 841 01 Bratislava, Slovak Republic
| |
Collapse
|
4
|
Fang L, Wang X, Zhang Y, Zhang C, Liu X, Li W, Zhang Y, Sun N, Zheng J, Wang G. Oncolytic Adenovirus Armoring with CXCL9 and IL15 Shows Potent Antitumor Activity and Boosts CAR-T Therapy for Prostate Cancer. Hum Gene Ther 2025. [PMID: 40346805 DOI: 10.1089/hum.2024.254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2025] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy has achieved great success and progress for treatment of hematological malignancy, but it still cannot overcome the obstacles in solid tumors. The hostile tumor microenvironment (TME), such as dense extracellular matrix, hypoxia, low pH, and tumor-derived metabolites, largely impedes CAR-T function. Oncolytic virus, as a form of immunotherapy, provides a way to antagonize the TME and improve the efficacy of CAR-T cells in solid tumors. In this study, the chemokine CXCL9 and interleukin 15 (IL15) genes were genetically integrated into adenoviral vector to construct oncolytic adenovirus (OAV) Ad-CXCL9-IL15, which could infect tumor cells to express and secrete CXCL9 and IL15. Ad-CXCL9-IL15 showed potent antitumor activity in xenografted prostate cancer model and augmented the tumor infiltration of CD45+CD3+ T and CD8+ T cells in immunocompetent mice. Moreover, Ad-CXCL9-IL15 treatment decreased Treg cells in tumor mass and increased CD44+CD62L+ T cells in spleen. Indicating that Ad-CXCL9-IL15 modified the TME and augmented antitumor immune responses in vivo. Furthermore, administration of Ad-CXCL9-IL15 dramatically promoted infiltration and survival of B7H3-targeting CAR-T cells, improved the therapeutic efficacy, and prolonged the survival time of prostate cancer-bearing mice. Therefore, cytokine-armored OAV Ad-CXCL9-IL15 could be used as a bioenhancer to modify TME and boost immunotherapy for solid tumors.
Collapse
Affiliation(s)
- Lin Fang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Xueyan Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Department of Oncology, Xuzhou City Hospital of TCM, Xuzhou, Jiangsu, China
| | - Yi Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Chen Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Department of Oncology, The First People's Hospital of Yancheng, Yancheng, Jiangsu, China
| | - Xiaoxiao Liu
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Wanjing Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Yuxin Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Nan Sun
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
5
|
Timpanaro A, Song EZ, Amwas N, Chiu CH, Ronsley R, Taylor MR, Foster JB, Wang LD, Vitanza NA. Evolving CAR T-Cell Therapy to Overcome the Barriers in Treating Pediatric Central Nervous System Tumors. Cancer Discov 2025; 15:890-902. [PMID: 40300089 PMCID: PMC12048232 DOI: 10.1158/2159-8290.cd-24-1465] [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: 10/09/2024] [Revised: 01/15/2025] [Accepted: 03/24/2025] [Indexed: 05/01/2025]
Abstract
SIGNIFICANCE CNS tumors are the leading cause of cancer-related death in children, highlighting the dire need for new treatment strategies. CAR T cells represent a unique approach, distinct from the cytotoxic chemotherapies and small-molecule inhibitors that have dominated the clinical trial space for decades. Phase I CAR T-cell trials have shown feasibility and possible efficacy against pediatric CNS tumors; however, many challenges must be overcome if these therapeutics are going to be beneficial to most affected children. Although rapid translational development and early-phase trials have quickly evolved our understanding, the pediatric CNS CAR T-cell community now yearns for critical assessments and open dialogue about overcoming the remaining obstacles ahead.
Collapse
Affiliation(s)
- Andrea Timpanaro
- Ben Towne Center for Childhood Cancer and Blood Disorders Research, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Edward Z. Song
- Ben Towne Center for Childhood Cancer and Blood Disorders Research, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Nour Amwas
- Department of Immuno-oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Chu-Hsuan Chiu
- Department of Immuno-oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Rebecca Ronsley
- Ben Towne Center for Childhood Cancer and Blood Disorders Research, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, Seattle Children’s Hospital, University of Washington, Seattle, WA, USA
| | - Mallory R. Taylor
- Ben Towne Center for Childhood Cancer and Blood Disorders Research, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, Seattle Children’s Hospital, University of Washington, Seattle, WA, USA
| | - Jessica B. Foster
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Leo D. Wang
- Department of Immuno-oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
- Department of Pediatrics, City of Hope Children’s Cancer Center, Duarte, CA, USA
| | - Nicholas A. Vitanza
- Ben Towne Center for Childhood Cancer and Blood Disorders Research, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, Seattle Children’s Hospital, University of Washington, Seattle, WA, USA
| |
Collapse
|
6
|
Liu R, Jiang X, Dong R, Zhang Y, Gai C, Wei P. Revealing the mechanisms and therapeutic potential of immune checkpoint proteins across diverse protein families. Front Immunol 2025; 16:1499663. [PMID: 40356928 PMCID: PMC12066663 DOI: 10.3389/fimmu.2025.1499663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 03/28/2025] [Indexed: 05/15/2025] Open
Abstract
Host immune responses to antigens are tightly regulated through the activation and inhibition of synergistic signaling networks that maintain homeostasis. Stimulatory checkpoint molecules initiate attacks on infected or tumor cells, while inhibitory molecules halt the immune response to prevent overreaction and self-injury. Multiple immune checkpoint proteins are grouped into families based on common structural domains or origins, yet the variability within and between these families remains largely unexplored. In this review, we discuss the current understanding of the mechanisms underlying the co-suppressive functions of CTLA-4, PD-1, and other prominent immune checkpoint pathways. Additionally, we examine the IgSF, PVR, TIM, SIRP, and TNF families, including key members such as TIGIT, LAG-3, VISTA, TIM-3, SIRPα, and OX40. We also highlight the unique dual role of VISTA and SIRPα in modulating immune responses under specific conditions, and explore potential immunotherapeutic pathways tailored to the distinct characteristics of different immune checkpoint proteins. These insights into the unique advantages of checkpoint proteins provide new directions for drug discovery, emphasizing that emerging immune checkpoint molecules could serve as targets for novel therapies in cancer, autoimmune diseases, infectious diseases, and transplant rejection.
Collapse
Affiliation(s)
| | | | | | | | - Cong Gai
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Peng Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
7
|
Zheng H, Tan J, Qin F, Zheng Y, Yang X, Liu Z, Cai W, Qin X, Liao H. PKM2 modulates chemotherapy sensitivity by regulating autophagy and predicts the prognosis and immunity in pancancer. Sci Rep 2025; 15:14626. [PMID: 40287473 PMCID: PMC12033356 DOI: 10.1038/s41598-025-96562-x] [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: 08/13/2024] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
One of the main characteristics of tumor metabolite reprogramming is enhanced glycolysis, and Pyruvate Kinase M2(PKM2) is a crucial enzyme that limits the pace of glycometabolism. Although PKM2 has been proven to affect the development of some cancers, a pan-cancer analysis of PKM2 has not yet been performed. We analyzed the expression and prognosis of PKM2 in pan-cancer using multiple databases. We performed epigenetic, functional enrichment, immune cell infiltration, immune checkpoint, and drug sensitivity analyses of PKM2. PKM2 was found to be significantly upregulated in most malignancies and associated with a bad prognosis. In some cancers, the PKM2 DNA promoter was hypomethylated. The expression of PKM2 was positively linked with most m6A-methylation-related genes in pan-cancer. The functions of PKM2 were primarily associated with the regulation of the immune system, glycolysis, hypoxia, angiogenesis, and epithelial-mesenchymal transition. PKM2 was favorably associated with neutrophils and cancer-associated fibroblasts in the tumor microenvironment of most cancers. Importantly, PKM2 showed a strikingly high correlation with CD274 (PD-L1), CD276, TGF-β1, VEGFA, and HAVCR2 in most cancers. Finally, using experiments, it was confirmed that silencing PKM2 could increase the sensitivity of esophageal squamous cell carcinoma to cisplatin by regulating autophagy. PKM2 affects autophagy - regulated tumor cell tolerance to chemotherapy, providing future research directions for solving tumor chemotherapy resistance.
Collapse
Affiliation(s)
- Haosheng Zheng
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Jian Tan
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Fei Qin
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Yuzhen Zheng
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Xingping Yang
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Zui Liu
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Weijie Cai
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Xianyu Qin
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
| | - Hongying Liao
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
| |
Collapse
|
8
|
Seo YR, Kim HB, Jung H, Kim EG, Huh S, Yi EC, Kim KM. Unveiling transcriptional mechanisms of B7-H3 in breast cancer stem cells through proteomic approaches. iScience 2025; 28:112218. [PMID: 40230524 PMCID: PMC11995042 DOI: 10.1016/j.isci.2025.112218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/29/2024] [Accepted: 03/11/2025] [Indexed: 04/16/2025] Open
Abstract
B7-H3, an immune checkpoint molecule, is prominently overexpressed in various solid tumors, correlating with poor clinical outcomes. Despite its critical role in promoting tumorigenesis, metastasis, and immune evasion, the regulatory mechanisms governing B7-H3 expression, particularly in cancer stem cells (CSCs), remain elusive. In this comprehensive study, we focused on breast CSCs to uncover the transcriptional regulators driving B7-H3 overexpression. Utilizing DNA affinity purification-mass spectrometry (DAP-MS) to analyze B7-H3 promoter regions, we identified a novel set of transcription factors, including DDB1, XRCC5, PARP1, RPA1, and RPA3, as key modulators of B7-H3 expression. Functional assays revealed that targeting DDB1 with nitazoxanide significantly downregulated B7-H3 expression, subsequently impairing tumor sphere formation and cell migration in breast CSCs. These findings not only elucidate the complex transcriptional network controlling B7-H3 expression but also open new avenues for developing targeted immunotherapies aimed at disrupting CSC-driven cancer progression.
Collapse
Affiliation(s)
- Yu Ri Seo
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- Institute of Medical and Biological Engineering, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Han Byeol Kim
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Hyeryeon Jung
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, Republic of Korea
| | - Eunhee G. Kim
- Department of Systems Immunology, Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Sumin Huh
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Eugene C. Yi
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- Institute of Medical and Biological Engineering, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Kristine M. Kim
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, Republic of Korea
- Department of Systems Immunology, Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
9
|
Xie Y, Wang X, Wang W, Pu N, Liu L. Epithelial-mesenchymal transition orchestrates tumor microenvironment: current perceptions and challenges. J Transl Med 2025; 23:386. [PMID: 40176117 PMCID: PMC11963649 DOI: 10.1186/s12967-025-06422-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/25/2025] [Indexed: 04/04/2025] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a critical process in cancer progression, facilitating tumor cells to develop invasive traits and augmenting their migratory capabilities. EMT is primed by tumor microenvironment (TME)-derived signals, whereupon cancer cells undergoing EMT in turn remodel the TME, thereby modulating tumor progression and therapeutic response. This review discusses the mechanisms by which EMT coordinates TME dynamics, including secretion of soluble factors, direct cell contact, release of exosomes and enzymes, as well as metabolic reprogramming. Recent evidence also indicates that cells undergoing EMT may differentiate into cancer-associated fibroblasts, thereby establishing themselves as functional constituents of the TME. Elucidating the relationship between EMT and the TME offers novel perspectives for therapeutic strategies to enhance cancer treatment efficacy. Although EMT-directed therapies present significant therapeutic potential, the current lack of effective targeting approaches-attributable to EMT complexity and its microenvironmental context dependency-underscores the necessity for mechanistic investigations and translational clinical validation.
Collapse
Affiliation(s)
- Yuqi Xie
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Xuan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenquan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ning Pu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
10
|
Mielcarska S, Kot A, Kula A, Dawidowicz M, Sobków P, Kłaczka D, Waniczek D, Świętochowska E. B7H3 in Gastrointestinal Tumors: Role in Immune Modulation and Cancer Progression: A Review of the Literature. Cells 2025; 14:530. [PMID: 40214484 PMCID: PMC11988818 DOI: 10.3390/cells14070530] [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: 03/11/2025] [Revised: 03/29/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
B7-H3 (CD276), a member of the B7 immune checkpoint family, plays a critical role in modulating immune responses and has emerged as a promising target in cancer therapy. It is highly expressed in various malignancies, where it promotes tumor evasion from T cell surveillance and contributes to cancer progression, metastasis, and therapeutic resistance, showing a correlation with the poor prognosis of patients. Although its receptors were not fully identified, B7-H3 signaling involves key intracellular pathways, including JAK/STAT, NF-κB, PI3K/Akt, and MAPK, driving processes crucial for supporting tumor growth such as cell proliferation, invasion, and apoptosis inhibition. Beyond immune modulation, B7-H3 influences cancer cell metabolism, angiogenesis, and epithelial-to-mesenchymal transition, further exacerbating tumor aggressiveness. The development of B7-H3-targeting therapies, including monoclonal antibodies, antibody-drug conjugates, and CAR-T cells, offers promising avenues for treatment. This review provides an up-to-date summary of the B7H3 mechanisms of action, putative receptors, and ongoing clinical trials evaluating therapies targeting B7H3, focusing on the molecule's role in gastrointestinal tumors.
Collapse
Affiliation(s)
- Sylwia Mielcarska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland; (A.K.); (P.S.); (D.K.)
| | - Anna Kot
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland; (A.K.); (P.S.); (D.K.)
| | - Agnieszka Kula
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-514 Katowice, Poland; (A.K.); (M.D.); (D.W.)
| | - Miriam Dawidowicz
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-514 Katowice, Poland; (A.K.); (M.D.); (D.W.)
| | - Piotr Sobków
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland; (A.K.); (P.S.); (D.K.)
| | - Daria Kłaczka
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland; (A.K.); (P.S.); (D.K.)
| | - Dariusz Waniczek
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-514 Katowice, Poland; (A.K.); (M.D.); (D.W.)
| | - Elżbieta Świętochowska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland; (A.K.); (P.S.); (D.K.)
| |
Collapse
|
11
|
Li XH, Xu JM. Dose-response relationship and predictive value of soluble B7-DC in bronchoalveolar lavage fluid and risk of refractory Mycoplasma pneumoniae pneumonia in children. Kaohsiung J Med Sci 2025; 41:e12944. [PMID: 39945309 DOI: 10.1002/kjm2.12944] [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: 10/18/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 04/02/2025] Open
Abstract
This study was to investigate the clinical significance of soluble B7-dendritic cell (sB7-DC) concentration in bronchoalveolar lavage fluid (BALF) of children with Refractory Mycoplasma pneumoniae pneumonia (RMPP). A total of 298 patients with Mycoplasma pneumoniae pneumonia (MPP) were enrolled. Patients were divided into general MPP (GMPP) (n = 213) and RMPP groups (n = 85). Detection of sB7-DC and serum inflammatory factors in BALF was performed by ELISA. The relationship between sB7-DC and the risk of RMPP was assessed using restricted cubic spline (RCS) model. A base model for predicting RMPP was constructed using logistic regression analysis, and a compound model was created with the addition of sB7-DC in the base model. ROC curves were plotted to evaluate the predictive value of the model. Column line plots were plotted to assess the contribution of each variable to the outcome event. Calibration curves were plotted and the Hosmer-Lemeshow test (HL test) was performed to assess the calibration performance of the model. Decision curve analysis (DCA) plots were plotted to assess determine whether sB7-DC has clinical value. There was no statistical difference between sB7-H3 and sB7-H4 in the two groups (both p > 0.05). sB7-DC levels were higher in the RMPP group than in the GMPP group (91.66 [77.36, 122.5] pg/ml vs. 64.87 [47.07, 86.46] pg/ml, p < 0.001). RCS analysis showed that the risk of RMPP gradually increased with the increase of sB7-DC when sB7-DC > 76.505 pg/ml. Both the base model and the compound model constructed with independent correlates of RMPP had some predictive value, and the models were well-fitted. The column line graphs showed that the models had discriminative ability. Notably, the compound model had a higher predictive value, with a higher AUC value than the base model: 0.76 (0.65-0.87) versus 0.68 (0.54-0.81). The highest net benefit was close to 0.15 (only 0.1 in the base model). When the net benefit was >0, the high-risk threshold took on a wide range of values. sB7-DC in children with RMPP is an independent predictor of RMPP. sB7-DC helps to improve quantitative prediction of RMPP risk and accurately guide medical decisions.
Collapse
Affiliation(s)
- Xue-Hua Li
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing City, China
| | - Jun-Mei Xu
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing City, China
| |
Collapse
|
12
|
Mielcarska S, Kula A, Dawidowicz M, Waniczek D, Świętochowska E. Prognostic Significance of B7H3 Expression in Solid Tumors: A Systematic Review and Meta-Analysis. Int J Mol Sci 2025; 26:3044. [PMID: 40243697 PMCID: PMC11988431 DOI: 10.3390/ijms26073044] [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: 01/13/2025] [Revised: 03/07/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
B7H3 (CD276), an immunoregulatory molecule known for its role in immune evasion by transmitting inhibitory signals to T lymphocytes, has garnered significant attention in recent years as a promising target for cancer immunotherapy. This interest is largely due to its high expression in various types of solid tumors, coupled with low protein levels in normal tissues. However, studies examining the impact of B7H3 on survival outcomes have shown inconsistent results, leaving its prognostic significance not fully clarified. Therefore, this meta-analysis aimed to assess the relationship between B7H3 expression and various prognostic parameters in patients with solid malignancies. PubMed, Web of Science (WOS), Cochrane, SCOPUS, and Embase databases were searched for eligible articles published until November 2024. Statistical analysis was performed using R studio (version 4.3.2). The analysis included a total of 51 eligible studies comprising 11,135 patients. Results showed that overexpression of B7H3 is a negative predictor for all examined survival outcomes: OS (HR = 1.71, 95% CI = 1.44-2.03, p < 0.0001), DFS (HR = 2.02, 95% CI = 1.49-2.73, p < 0.0001), PFS (HR = 2.10, 95% CI = 1.44-3.06, p < 0.0001), RFS (HR = 1.66, 95% CI = 1.11-2.48, p = 0.01), and DSS (HR = 1.70, 95% CI = 1.24-2.32, p < 0.01). Despite the high heterogeneity observed across the studies, the sensitivity analysis confirmed the robustness of these results. This research suggests that B7H3 may serve as an effective biomarker for prognosis in solid tumors.
Collapse
Affiliation(s)
- Sylwia Mielcarska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-800 Zabrze, Poland
| | - Agnieszka Kula
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-514 Katowice, Poland; (A.K.); (M.D.); (D.W.)
| | - Miriam Dawidowicz
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-514 Katowice, Poland; (A.K.); (M.D.); (D.W.)
| | - Dariusz Waniczek
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-514 Katowice, Poland; (A.K.); (M.D.); (D.W.)
| | - Elżbieta Świętochowska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-800 Zabrze, Poland
| |
Collapse
|
13
|
Wen J, Wu X, Shu Z, Wu D, Yin Z, Chen M, Luo K, Liu K, Shen Y, Le Y, Shu Q. Clusterin-mediated polarization of M2 macrophages: a mechanism of temozolomide resistance in glioblastoma stem cells. Stem Cell Res Ther 2025; 16:146. [PMID: 40128761 PMCID: PMC11934612 DOI: 10.1186/s13287-025-04247-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 02/20/2025] [Indexed: 03/26/2025] Open
Abstract
Glioblastoma remains one of the most lethal malignancies, largely due to its resistance to standard chemotherapy such as temozolomide. This study investigates a novel resistance mechanism involving glioblastoma stem cells (GSCs) and the polarization of M2-type macrophages, mediated by the extracellular vesicle (EV)-based transfer of Clusterin. Using 6-week-old male CD34+ humanized huHSC-(M-NSG) mice (NM-NSG-017) and glioblastoma cell lines (T98G and U251), we demonstrated that GSC-derived EVs enriched with Clusterin induce M2 macrophage polarization, thereby enhancing temozolomide resistance in glioblastoma cells. Single-cell and transcriptome sequencing revealed close interactions between GSCs and M2 macrophages, highlighting Clusterin as a key mediator. Our findings indicate that Clusterin-rich EVs from GSCs drive glioblastoma cell proliferation and resistance to temozolomide by modulating macrophage phenotypes. Targeting this pathway could potentially reverse resistance mechanisms, offering a promising therapeutic approach for glioblastoma. This study not only sheds light on a critical pathway underpinning glioblastoma resistance but also lays the groundwork for developing therapies targeting the tumor microenvironment. Our results suggest a paradigm shift in understanding glioblastoma resistance, emphasizing the therapeutic potential of disrupting EV-mediated communication in the tumor microenvironment.
Collapse
Affiliation(s)
- Jianping Wen
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China.
| | - Xia Wu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China
| | - Zhicheng Shu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China
| | - Dongxu Wu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China
| | - Zonghua Yin
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China
| | - Minglong Chen
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China
| | - Kun Luo
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China
| | - Kebo Liu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China
| | - Yulong Shen
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China
| | - Yi Le
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China
| | - Qingxia Shu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China.
| |
Collapse
|
14
|
Du Q, Meng C, Zhang W, Huang L, Xue C. Establishing a Prognostic Model Correlates to Inflammatory Response Pathways for Prostate Cancer via Multiomic Analysis of Lactylation-Related Genes. Int J Genomics 2025; 2025:6681711. [PMID: 40161494 PMCID: PMC11952923 DOI: 10.1155/ijog/6681711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Prostate cancer (PCa) continues to pose substantial clinical challenges, with molecular heterogeneity significantly impacting therapeutic decision-making and disease trajectories. Emerging evidence implicates protein lactylation-a novel epigenetic regulatory mechanism-in oncogenic processes, though its prognostic relevance in PCa remains underexplored. Through integrative bioinformatics interrogation of lactylation-associated molecular signatures, we established prognostic correlations using multivariable feature selection methodologies. Initial screening via differential expression analysis (limma package) coupled with Cox proportional hazards modeling revealed 11 survival-favorable regulators and 16 hazard-associated elements significantly linked to biochemical recurrence. To enhance predictive precision, ensemble machine learning frameworks were implemented, culminating in a 10-gene lactylation signature demonstrating robust discriminative capacity (concordance index = 0.738) across both primary (TCGA-PRAD) and external validation cohorts (DKFZ). Multivariable regression confirmed the lactylation score's prognostic independence, exhibiting prominent associations with clinicopathological parameters including tumor staging and metastatic potential. The developed clinical-molecular nomogram achieved superior predictive accuracy (C - index > 0.7) through the synergistic integration of biological and clinical covariates. Tumor microenvironment deconvolution uncovered distinct immunological landscapes, with high-risk stratification correlating with enriched stromal infiltration and immunosuppressive phenotypes. Pathway enrichment analyses implicated chromatin remodeling processes and cytokine-mediated inflammatory cascades as potential mechanistic drivers of prognostic divergence. Therapeutic vulnerability profiling demonstrated differential response patterns: low-risk patients exhibited enhanced immune checkpoint inhibitor responsiveness, whereas high-risk subgroups showed selective chemosensitivity to docetaxel and mitoxantrone. Functional validation in PC-3 models revealed AK5 silencing induced proapoptotic effects, suppressed metastatic potential of migration and invasion, and modulated immune checkpoint regulation through CD276 coexpression. These multimodal findings position lactylation dynamics, particularly AK5-mediated pathways, as promising therapeutic targets and stratification biomarkers in PCa management.
Collapse
Affiliation(s)
- Qinglong Du
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - CuiYu Meng
- The Department of EICU, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong, China
| | - Wenchao Zhang
- The Department of Urology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong, China
| | - Li Huang
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, Guangdong, China
| | - Chunlei Xue
- The Department of Urology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong, China
| |
Collapse
|
15
|
Tolmachev V, Papalanis E, Bezverkhniaia EA, Rosly AH, Vorobyeva A, Orlova A, Carlqvist M, Frejd FY, Oroujeni M. Impact of Radiometal Chelates on In Vivo Visualization of Immune Checkpoint Protein Using Radiolabeled Affibody Molecules. ACS Pharmacol Transl Sci 2025; 8:706-717. [PMID: 40109742 PMCID: PMC11915182 DOI: 10.1021/acsptsci.4c00539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 03/22/2025]
Abstract
The immune checkpoint protein B7-H3 (CD276) is overexpressed in various cancers and is an attractive target for the treatment of malignant tumors. Radionuclide molecular imaging of B7-H3 expression using engineered scaffold proteins such as Affibody molecules is a promising strategy for the selection of potential responders to B7-H3-targeted therapy. Feasibility of B7-H3 imaging was demonstrated using two 99mTc-labeled probes, AC12 and an affinity-matured SYNT179 using a [99mTc]Tc-GGGC label. This study aimed to evaluate whether the use of a residualizing 111In-based label provides better imaging contrast compared with a nonresidualizing label. To do that, SYNT179 and AC12-GGGC Affibody molecules were labeled with 111In using (4,10-bis-carboxymethyl-7-{[2-(2,5-dioxo-3-thioxo-pyrrolidin-1-yl)-ethylcarbamoyl]-methyl}-1,4,7,10-tetraaza-cyclododec-1-yl)-acetic acid (maleimide-DOTA) chelator, site-specifically coupled to the C-terminus of Affibody molecules. The binding affinities of the 111In-labeled conjugates to B7-H3-expressing living cells were higher compared with the affinities of the 99mTc-labeled variants. In mice with B7-H3-expressing xenografts, the tumor uptake of 111In-labeled proteins (3.6 ± 0.3 and 1.8 ± 0.5%ID/g for [111In]In-SYNT179-DOTA and [111In]In-AC12-DOTA, respectively) was significantly (p < 0.05, ANOVA) higher than those for 99mTc-labeled counterparts (1.6 ± 0.2%ID/g and 0.8 ± 0.2%ID/g for [99mTc]Tc-SYNT179 and [99mTc]Tc-AC12-GGGC, respectively). The best variant, [111In]In-SYNT179-DOTA, provided a tumor-to-blood ratio of 31.1 ± 2.9, which was twice higher than that for [99mTc]Tc-SYNT179 and 7-fold higher than that for [99mTc]Tc-AC12-GGGC. Both 111In-labeled Affibody molecules had higher renal retention compared with 99mTc-labeled ones, but the hepatobiliary excretion of 111In-labeled proteins was appreciably lower, potentially improving the imaging of abdominal metastases. Overall, [111In]In-SYNT179-DOTA is the most promising tracer for visualization of B7-H3 expression.
Collapse
Affiliation(s)
- Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Eleftherios Papalanis
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | | | - Alia Hani Rosly
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 751 83 Uppsala, Sweden
| | | | - Fredrik Y Frejd
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
- Affibody AB, 171 65 Solna, Sweden
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| |
Collapse
|
16
|
Che J, Liu Y, Liu Y, Song J, Cui H, Feng D, Tian A, Zhang Z, Xu Y. The application of emerging immunotherapy in the treatment of prostate cancer: progress, dilemma and promise. Front Immunol 2025; 16:1544882. [PMID: 40145100 PMCID: PMC11937122 DOI: 10.3389/fimmu.2025.1544882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
In recent years, there has been a growing trend towards the utilization of immunotherapy techniques for the treatment of cancer. Some malignancies have acquired significant progress with the use of cancer vaccines, immune checkpoint inhibitors, and adoptive cells therapy. Scholars are exploring the aforementioned methods as potential treatments for advanced prostate cancer (PCa) due to the absence of effective adjuvant therapy to improve the prognosis of metastatic castration-resistant prostate cancer (mCRPC). Immunotherapy strategies have yet to achieve significant advancements in the treatment of PCa, largely attributed to the inhibitory tumor microenvironment and low mutation load characteristic of this malignancy. Hence, researchers endeavor to address these challenges by optimizing the design and efficacy of immunotherapy approaches, as well as integrating them with other therapeutic modalities. To date, studies have also shown potential clinical benefits. This comprehensive review analyzed the utilization of immunotherapy techniques in the treatment of PCa, assessing their advantages and obstacles, with the aim of providing healthcare professionals and scholars with a comprehensive understanding of the progress in this field.
Collapse
Affiliation(s)
- Jizhong Che
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Yuanyuan Liu
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Yangyang Liu
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Jingheng Song
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Hongguo Cui
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Dongdong Feng
- Department of Urology, Haiyang City People’s Hospital, Yantai, Shandong, China
| | - Aimin Tian
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Zhengchao Zhang
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Yankai Xu
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
17
|
Tamura Y, Ohki S, Nagai H, Yoshizato R, Nishi S, Jin Y, Kitajima Y, Guo Y, Ichinohe T, Okada S, Kawano Y, Yasuda T. Co-expression of B7-H3 and LAG3 represents cytotoxicity of CD4 + T cells in humans. Front Immunol 2025; 16:1560383. [PMID: 40070836 PMCID: PMC11893609 DOI: 10.3389/fimmu.2025.1560383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Recent studies have highlighted the potential contribution of CD4+ T cells with cytotoxic activity (CD4 CTLs) to anti-tumor immunity. However, their precise roles remain elusive, partly due to the absence of specific markers defining CD4 CTLs with target-killing potential in humans. We previously demonstrated that Epstein-Barr virus (EBV)-driven immortalized B cell lines efficiently induce human CD4 CTLs with cytotoxic functions comparable to cytotoxic CD8+ T cells (CD8 CTLs). Here we show that EBV-driven CD4 CTLs exhibit prolonged proliferation and sustained cytotoxicity compared with CD8 CTLs, although their cytotoxic function markedly decreased during long-term culture. Comparative transcriptomic analysis of CD4 CTLs with varying cytotoxic activities identified B7-H3 and LAG3 as surface molecules associated with highly cytotoxic CD4 CTLs. Co-expression of B7-H3 and LAG3 correlated with CD107a expression and was observed on CD4+ T cells with enhanced cytotoxic potential in a target-dependent manner but not on CD8 CTLs. Furthermore, B7-H3+LAG3+ CD4+ T cells were induced during co-culture with bone marrow cells from pediatric patients with B-cell acute lymphoblastic leukemia (B-ALL). These findings suggest that B7-H3 and LAG3 co-expression represents a characteristic feature of functional CD4 CTLs in humans, providing valuable insights into the role of CD4 CTLs in tumor immunity.
Collapse
Affiliation(s)
- Yumi Tamura
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shun Ohki
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Haruna Nagai
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Rin Yoshizato
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shizuki Nishi
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuqi Jin
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yasuo Kitajima
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yun Guo
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tatsuo Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Satoshi Okada
- Department of Pediatrics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yohei Kawano
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomoharu Yasuda
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
18
|
Yüceer RO, Kaya S, Balcı SN, Eğilmez HR, Yılmaz M, Erdıs E. Prognostic Biomarkers in Isocitrate Dehydrogenase Wild-Type Glioblastoma: A Focus on B7-H3. Brain Sci 2025; 15:212. [PMID: 40002543 PMCID: PMC11853153 DOI: 10.3390/brainsci15020212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Isocitrate dehydrogenase (IDH) wild-type (wt) glioblastoma is an aggressive malignancy associated with poor clinical outcomes, marked by high heterogeneity and resistance to treatment. This study aims to investigate the prognostic significance of B7-H3 expression in IDH wt glioblastoma and its potential association with clinical outcomes, including overall survival (OS) and progression-free survival (PFS). Additionally, the relationship between B7-H3 and PD-L1 expression was explored. METHODS A retrospective cohort of 86 IDH wt glioblastoma patients, all of whom underwent surgery, radiotherapy, and temozolomide treatment, was analyzed. B7-H3 expression was quantified using an immunoreactivity score (IRS), classifying samples as low (IRS ≤ 4) or high (IRS > 4). PD-L1 expression was evaluated based on tumor and immune cell staining, with >5% positivity indicating significant expression. RESULTS High B7-H3 expression was significantly associated with poorer OS and PFS. Co-expression of B7-H3 and PD-L1 was prevalent, particularly among younger male patients with unifocal tumors; however, PD-L1 expression did not show a significant correlation with clinical outcomes. CONCLUSIONS B7-H3 appears to be a promising prognostic biomarker in IDH wt glioblastoma and may serve as a target for developing combination therapies, integrating B7-H3-targeting treatments with immune checkpoint inhibitors. Further prospective studies are necessary to validate these findings and to explore potential therapeutic strategies.
Collapse
Affiliation(s)
- Ramazan Oğuz Yüceer
- Department of Pathology, Sivas Cumhuriyet University School of Medicine, 58140 Sivas, Turkey; (S.K.); (H.R.E.)
| | - Seyhmus Kaya
- Department of Pathology, Sivas Cumhuriyet University School of Medicine, 58140 Sivas, Turkey; (S.K.); (H.R.E.)
| | - Sema Nur Balcı
- Sivas Cumhuriyet University School of Medicine, 58140 Sivas, Turkey;
| | - Hatice Reyhan Eğilmez
- Department of Pathology, Sivas Cumhuriyet University School of Medicine, 58140 Sivas, Turkey; (S.K.); (H.R.E.)
| | - Mukaddes Yılmaz
- Department of Clinical Oncology, Sivas Cumhuriyet University School of Medicine, 58140 Sivas, Turkey;
| | - Eda Erdıs
- Department of Radiation Oncology, Sivas Cumhuriyet University School of Medicine, 58140 Sivas, Turkey;
| |
Collapse
|
19
|
Feigelson SW, Dadosh T, Levi N, Sapoznikov A, Weinstein-Marom H, Blokon-Kogan D, Avraham Y, Unger T, Gross G, Dahan R, Alon R. CD32B1, a versatile non-signaling antibody-binding scaffold for enhanced T cell adhesion to tumor stromal cognate antigens. Front Immunol 2025; 16:1398757. [PMID: 39995660 PMCID: PMC11847833 DOI: 10.3389/fimmu.2025.1398757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 01/15/2025] [Indexed: 02/26/2025] Open
Abstract
Targeting cytotoxic T lymphocytes (CTLs), as chimeric antigen T cells (CAR-T), T cell receptor-engineered (TCR)-T cells or adoptive cell transfer of tumor infiltrating T cells (TILs) to solid tumors is a major therapeutic challenge. We describe a new strategy to confer these lymphocytes with de novo adhesiveness to surface proteins enriched in the tumor microenvironment. This approach is based on decorating CTLs with monoclonal antibodies (mAbs) specific to any surface protein of interest within the stroma and the extracelullar matrix of solid tumors. For efficient mAb decoration, we have introduced a mAb binding Fc receptor (FcR) scaffold, FcγRIIB1 (CD32B1), which we found to be enriched on B lymphocyte microvilli (MV). This isoform contains an inhibitory ITIM motif within a cytoplasmic tail anchored to the cortical cytoskeleton. We thus generated a non-signaling CD32B1 mutant lacking the ITIM motif (termed ITIM-less CD32B1, or ILCD32B1) and successfully expressed it in human T cells which normally do not express this FcR. The ILCD32B1 expressing lymphocytes bound multiple IgG1 mAbs whose Fc domain was engineered with a 5-residue substitution to reach a nM range of Fc-FcγCR dissociation constants. The mAb decorated ILCD32B1 expressing T cells could readily adhere to a surface-bound cognate antigen. To broaden the utility of this scaffold, we have also generated a new fusion protein in which the entire Fc binding domain was truncated (tILCD32B1) and replaced with a monomeric streptavidin variant, mSA2, via a CD8 hinge. The molecule, termed mSA2-CD8h-tILCD32B1, was also successfully expressed in T cells, readily and stably bound biotinylated IgG mAbs in vitro and once decorated with the biotin labeled mAbs, conferred the T cells with high adhesiveness to multiple surface-coated antigens. mSA2-CD8h-tILCD32B1 expressing human T cells decorated ex vivo with a biotin-labeled mAb retained the antibody for hours after accumulation inside breast tumors implanted in immunodeficient recipient mice. Our results collectively suggest that a non-signaling CD32B1 can be used as a versatile scaffold for mAb decoration of T cells. Our mAb decoration approach can confer new cell adhesive reactivities to improve tumor CTL (CAR-T and TIL) accumulation and retention inside solid tumors.
Collapse
Affiliation(s)
- Sara W. Feigelson
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Tali Dadosh
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Nehora Levi
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Anita Sapoznikov
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hadas Weinstein-Marom
- Laboratory of Immunology, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel
- Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel
| | - Dayana Blokon-Kogan
- Laboratory of Immunology, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel
- Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel
| | - Yahel Avraham
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Tamar Unger
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Gideon Gross
- Laboratory of Immunology, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel
- Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel
| | - Rony Dahan
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Ronen Alon
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
20
|
Shi LL, Chen Y, Xie MX, Chen QZ, Qiao XW, Cheng QH, Li L, Fu R, Liang T, Jiang X, Wang MJ, Yao J, Li JJ. UBE2T/CDC42/CD276 signaling axis mediates brain metastasis of triple-negative breast cancer via lysosomal autophagy. J Immunother Cancer 2025; 13:e010782. [PMID: 39915000 PMCID: PMC11804199 DOI: 10.1136/jitc-2024-010782] [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: 10/11/2024] [Accepted: 01/26/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Advanced triple-negative breast cancer (TNBC) is prone to brain metastasis (BrM). The precise molecular mechanism responsible for this phenomenon has not yet been completely established, so it is vital to comprehend the molecular mechanism behind it. METHODS The protein chip analysis was conducted to identify any abnormal UBE2T protein expression in TNBC, especially BrM. Here, we used public databases and bioinformatics analysis as well as clinical samples from different cohorts to investigate the interrelationship between UBE2T/CDC42/CD276. This predicted relationship was then repeatedly validated using different in vivo and in vitro experimental methods. Additionally, multiple experimental approaches were implemented, encompassing western blotting, Co-IP, GST pull-down, flow cytometry, mass spectrometry, immunofluorescence, immunohistochemistry, and qRT-PCR to reveal the molecular mechanism of UBE2T-mediated immune escape and BrM. RESULTS Our results indicate that expressed at elevated levels in breast cancer, UBE2T is negatively linked to patient prognosis, especially in BrM of TNBC. Data from clinical samples from our different cohorts and TCGA indicate a significant correlation between UBE2T and immunosuppression. Mechanistically, UBE2T directly interacts with CDC42, promoting its K48-linked polyubiquitination and proteasomal degradation, thereby inhibiting CDC42 from degrading CD276 via the autophagy-lysosomal pathway, indirectly upregulating CD276 and thereby impairing the CD8+ T cells function, ultimately mediating tumor immune escape and BrM. Finally, animal experimental results also showed that inhibition of UBE2T elevated the TNBC sensitivity to immune checkpoint CD276 blockade and inhibited BrM of TNBC. CONCLUSIONS In conclusion, our results indicate a new mechanism whereby UBE2T-mediated ubiquitination positively controls the UBE2T/CDC42/CD276 axis to upregulate tumor cell expression of CD276 and thereby impair CD8+ T cells function, ultimately leading to tumor cell immune escape and BrM.
Collapse
Affiliation(s)
| | - Yan Chen
- Department of Ultrasound Medicine, Wuhan Union Hospital, Wuhan, Hubei, China
| | - Ming Xing Xie
- Department of Ultrasound Medicine, Wuhan Union Hospital, Wuhan, Hubei, China
| | - Qian Zhi Chen
- Department of Breast and Thyroid Surgery, Wuhan Union Hospital, Wuhan, Hubei, China
| | - Xin Wei Qiao
- Department of Thoracic Surgery, Wuhan Union Hospital, Wuhan, Hubei, China
| | - Qi Hong Cheng
- Department of Neurosurgery, Wuhan Union Hospital, Wuhan, Hubei, China
| | - Lin Li
- Department of Neurosurgery, Wuhan Union Hospital, Wuhan, Hubei, China
| | - Rong Fu
- Department of Neurosurgery, Wuhan Union Hospital, Wuhan, Hubei, China
| | - Tao Liang
- Department of Clinical Laboratory, Wuhan Union Hospital, Wuhan, Hubei, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Wuhan Union Hospital, Wuhan, Hubei, China
| | - Min Jie Wang
- Department of Neurosurgery, Wuhan Union Hospital, Wuhan, Hubei, China
| | - Jin Yao
- Cancer Center, Wuhan Union Hospital, Wuhan, Hubei, China
| | - Jun Jun Li
- Department of Neurosurgery, Wuhan Union Hospital, Wuhan, Hubei, China
| |
Collapse
|
21
|
Haberecker M, Rüschoff JH, Andriakopoulou C, Gray SG, Nackaerts K, De Perrot M, Brcic L, Nadal E, Tsimpoukis S, Ampollini L, Aerts JG, Kirschner MB, Monkhorst K, Weynand B, Bavaghar-Zaeimi F, Samarzija M, Llatjos R, Finn SP, Silini E, Von Der Thüsen J, Vagenknecht P, Tsourti Z, Kerr KM, Kammler R, Peters S, Baas P, Opitz I, Stahel RA, Curioni-Fontecedro A. Prevalence and Clinical Association of CD276 (B7-H3) Expression in Pleural Mesothelioma: Results From the European Thoracic Platform Mesoscape Project. JCO Precis Oncol 2025; 9:e2400675. [PMID: 39938010 DOI: 10.1200/po-24-00675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/26/2024] [Accepted: 01/08/2025] [Indexed: 02/14/2025] Open
Abstract
PURPOSE CD276 (B7-H3) is an immunoregulatory protein that plays an important role in the inhibition of T-cell function. CD276 is overexpressed on a variety of human solid cancer cells with limited expression in normal tissues, making it an appealing target for innovative cancer immunotherapy approaches. Pleural mesothelioma (PM) is a highly aggressive disease with a need for new treatment options. Our objective was to investigate the expression of CD276 in the multicenter PM cohort of the European Thoracic Oncology Platform Mesoscape project and correlate the results with annotated clinical data. MATERIALS AND METHODS Using tissue microarrays (TMAs), the expression of CD276, assessed using a semiquantitative aggregate H-score method on the membrane (and secondarily in the cytoplasm), was correlated with clinicopathologic characteristics and survival outcome. RESULTS CD276 immunohistochemistry results were available for 353 patients, with mostly epithelioid histology (71%). Membranous CD276 expression was present in 86%. High membranous CD276 expression (H-score ≥the median H-score of 120) was significantly more common in females (P = .0029; 71% v 47%) and in epithelioid histology (P < .001; 59% v 29%), whereas no significant association in clinical outcome (overall survival [OS]/progression-free survival) was found. Cross-validation of the TMA method using whole sections revealed a moderate agreement for membranous assessment (Cohen's kappa = 0.47) and a lower agreement for cytoplasm assessment (Cohen's kappa = 0.37). In an exploratory analysis, high cytoplasmic CD276 expression was associated with worse prognosis (OS, log-rank P = .043), but was not significant when adjusting for other clinical variables. CONCLUSION Although no prognostic value of CD276 expression was found, its high membranous expression (86%) in the PM samples of the study supports further research of its potential as a therapeutic target for this disease.
Collapse
Affiliation(s)
- Martina Haberecker
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Jan H Rüschoff
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Charitini Andriakopoulou
- ETOP Statistical Office, ETOP IBCSG Partners Foundation, Frontier Science Foundation-Hellas, Athens, Greece
| | - Steven G Gray
- Department of Clinical Medicine, St James's Hospital and Trinity College Dublin, Dublin, Ireland
| | - Kristiaan Nackaerts
- Department of Respiratory Oncology, KU Leuven-University Hospital Leuven, Leuven, Belgium
| | - Marc De Perrot
- Division of Thoracic Surgery, Toronto General Hospital, Toronto, Canada
| | - Luka Brcic
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Gdansk, Austria
| | - Ernest Nadal
- Department of Medical Oncology, Catalan Institute of Oncology, IDIBELL, L'Hospitalet, Barcelona, Spain
| | - Sotirios Tsimpoukis
- Medical School of Athens, National and Kapodistrian University, Sotiria General Hospital, Athens, Greece
| | - Luca Ampollini
- Thoracic Surgery, Department of Medicine and Surgery, University Hospital of Parma, Parma, Italy
| | - Joachim G Aerts
- Thoracic Oncology Department, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Kim Monkhorst
- Division of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Birgit Weynand
- Department of Imaging and Pathology, University Hospitals Leuven, Leuven, Belgium
| | | | - Miroslav Samarzija
- Department for Lung Diseases, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Roger Llatjos
- Department of Pathology, Hospital Universitari de Bellvitge, L'Hospitalet, Barcelona, Spain
| | - Stephen P Finn
- Cancer Molecular Diagnostics and Histopathology, St James's Hospital and Trinity College Dublin, Dublin, Ireland
| | - Enrico Silini
- Thoracic Surgery, Department of Medicine and Surgery, University Hospital of Parma, Parma, Italy
| | - Jan Von Der Thüsen
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Patrick Vagenknecht
- Translational Research Coordination, ETOP IBCSG Partners Foundation, Bern, Switzerland
| | - Zoi Tsourti
- ETOP Statistical Office, ETOP IBCSG Partners Foundation, Frontier Science Foundation-Hellas, Athens, Greece
| | - Keith M Kerr
- Department of Pathology, Aberdeen Royal Infirmary-NHS Grampian, Aberdeen, United Kingdom
| | - Roswitha Kammler
- Translational Research Coordination, ETOP IBCSG Partners Foundation, Bern, Switzerland
| | - Solange Peters
- Department of Oncology, CHUV, Lausanne University Hospital, Lausanne, Switzerland
| | - Paul Baas
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Isabelle Opitz
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
22
|
Sang M, Ge J, Ge J, Tang G, Wang Q, Wu J, Mao L, Ding X, Zhou X. Immune regulatory genes impact the hot/cold tumor microenvironment, affecting cancer treatment and patient outcomes. Front Immunol 2025; 15:1382842. [PMID: 39911580 PMCID: PMC11794490 DOI: 10.3389/fimmu.2024.1382842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 12/31/2024] [Indexed: 02/07/2025] Open
Abstract
Background and aims Immunologically hot tumors, characterized by an inflamed tumor microenvironment (TME), contrast significantly with immunologically cold tumors. The identification of these tumor immune subtypes holds clinical significance, as hot tumors may exhibit improved prognoses and heightened responsiveness to checkpoint blockade therapy. Nevertheless, as yet there is no consensus regarding the clinically relevant definition of hot/cold tumors, and the influence of immune genes on the formation of hot/cold tumors remains poorly understood. Methods Data for 33 different types of cancer were obtained from The Cancer Genome Atlas database, and their immune composition was assessed using the CIBERSORT algorithm. Tumors were categorized as either hot or cold based on their distinct immune composition, ongoing immune response, and overall survival. A customized immunogram was created to identify important immunological characteristics. Kyoto Encyclopedia of Genes and Genomes and Hallmark pathway enrichment were evaluated through gene set variation analysis. Additionally, hub genes that regulate the tumor microenvironment were identified, and their expression patterns were analyzed using single-cell RNA sequencing. Furthermore, drug sensitivity and molecular docking analyses were performed to identify potential drug candidates capable of transforming cold tumors into hot tumors. For validation, a clinical cohort of patients diagnosed with pancreatic adenocarcinoma was examined using multiplex immunohistochemistry. Results We were able to differentiate between hot and cold tumors in various types of cancer (bladder urothelial carcinoma, pancreatic adenocarcinoma, and cervical squamous cell carcinoma) by analyzing the presence of CD8+ T cells, activated natural killer cells, and M2-type macrophages, as well as the cytolytic activity and T cell proliferation. Hub genes that regulate the TME, including PDCD1, CD276, and NT5E, were discovered. The increased expression of NT5E and its prognostic significance were confirmed through multiplex immunohistochemistry in pancreatic adenocarcinoma. Finally, dasatinib and tozasertib were identified as drug candidates capable of converting cold pancreatic adenocarcinoma tumors into hot tumors. Conclusion In this study, we developed a framework for discerning clinically significant immune subtypes across various cancer types, further identifying several potential targets for converting cold tumors into hot tumors to enhance anticancer treatment efficacy.
Collapse
Affiliation(s)
- Mengmeng Sang
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Jia Ge
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Juan Ge
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
- Department of Respiratory Medicine, Affiliated Nantong Hospital of Shanghai University, Nantong, China
| | - Gu Tang
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Qiwen Wang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jiarun Wu
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Liming Mao
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
- Basic Medical Research Center, School of Medicine, Nantong University, Nantong, China
| | - Xiaoling Ding
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| |
Collapse
|
23
|
Ma Z, Yang J, Jia W, Li L, Li Y, Hu J, Luo W, Li R, Ye D, Lan P. Histone lactylation-driven B7-H3 expression promotes tumor immune evasion. Theranostics 2025; 15:2338-2359. [PMID: 39990209 PMCID: PMC11840737 DOI: 10.7150/thno.105947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/05/2025] [Indexed: 02/25/2025] Open
Abstract
Rationale: Tumor cells possess sophisticated strategies to circumvent immune detection, including the modulation of endogenous immune checkpoints, particularly those within the B7 family. Elucidating the mechanisms that govern the induction of B7 family molecules is crucial for the advancement of immunotherapy. Lysine lactylation (Kla), a newly identified epigenetic modification, is suggested may play a role in reshaping the tumor microenvironment and facilitating immune evasion. Methods: We analyzed the glycolysis pathway's enrichment in patients with immune-evading tumors and assessed the impact of lactate treatment on the antitumor immunity of CD8+ T cells in the tumor microenvironment. We interrupted glycolysis using lactate dehydrogenase A (LDHA) knockdown and sodium oxamate, and evaluated its effects on CD8+ T cell cytotoxicity. Additionally, we investigated the correlation between B7-H3 expression and the glycolysis pathway, and explored the molecular mechanisms underlying lactate-induced B7-H3 expression. Results: Our findings revealed that the glycolysis pathway was highly enriched in immune-evading tumors. Lactate treatment inhibited the antitumor immunity of CD8+ T cells, whereas interruption of glycolysis via LDHA knockdown or treatment with sodium oxamate augmented the cytotoxicity of CD8+ T cells, effectively counteracting tumor immune evasion. B7-H3 expression was found to be closely linked with the glycolysis pathway. Mechanistically, lactate-upregulated H3K18la directly bound to the B7-H3 promoter in conjunction with the transcription factor Creb1 and its co-activator Ep300, leading to increased B7-H3 expression and contributing to tumor progression by compromising the proportion and cytotoxicity of tumor-infiltrating CD8+ T cells. In mouse tumor bearing models, inhibiting glycolysis and B7-H3 expression suppressed tumor cell growth, activated tumor-infiltrating CD8+ T cells, and demonstrated potent anti-tumor efficacy. Furthermore, this approach enhanced the efficacy of anti-PD-1 treatment. Conclusions: This study uncovers a novel mechanism by which lactate modulates the immune microenvironment through the glycolysis pathway and B7-H3 expression, providing new avenues for lactate metabolism-targeted tumor immunotherapy.
Collapse
Affiliation(s)
- Zhibo Ma
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China
| | - Jincui Yang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China
- Department of oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wenlong Jia
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Le Li
- Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yixin Li
- Department of oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Junjie Hu
- Department of oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Luo
- Department of oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ronghui Li
- Department of neurosurgery, Affiliated Hospital of Shandong University of traditional Chinese Medicine, Weifang, 250100, China
| | - Dawei Ye
- Department of oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Peixiang Lan
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, 430030 Wuhan, People's Republic of China
| |
Collapse
|
24
|
Khan SH, Choi Y, Veena M, Lee JK, Shin DS. Advances in CAR T cell therapy: antigen selection, modifications, and current trials for solid tumors. Front Immunol 2025; 15:1489827. [PMID: 39835140 PMCID: PMC11743624 DOI: 10.3389/fimmu.2024.1489827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 12/02/2024] [Indexed: 01/22/2025] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has revolutionized the treatment of hematologic malignancies, achieving remarkable clinical success with FDA-approved therapies targeting CD19 and BCMA. However, the extension of these successes to solid tumors remains limited due to several intrinsic challenges, including antigen heterogeneity and immunosuppressive tumor microenvironments. In this review, we provide a comprehensive overview of recent advances in CAR T cell therapy aimed at overcoming these obstacles. We discuss the importance of antigen identification by emphasizing the identification of tumor-specific and tumor-associated antigens and the development of CAR T therapies targeting these antigens. Furthermore, we highlight key structural innovations, including cytokine-armored CARs, protease-regulated CARs, and CARs engineered with chemokine receptors, to enhance tumor infiltration and activity within the immunosuppressive microenvironment. Additionally, novel manufacturing approaches, such as the Sleeping Beauty transposon system, mRNA-based CAR transfection, and in vivo CAR T cell production, are discussed as scalable solution to improve the accessibility of CAR T cell therapies. Finally, we address critical therapeutic limitations, including cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and suboptimal persistence of CAR T cells. An examination of emerging strategies for countering these limitations reveals that CRISPR-Cas9-mediated genetic modifications and combination therapies utilizing checkpoint inhibitors can improve CAR T cell functionality and durability. By integrating insights from preclinical models, clinical trials, and innovative engineering approaches, this review addresses advances in CAR T cell therapies and their performance in solid tumors.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Neoplasms/therapy
- Neoplasms/immunology
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Antigens, Neoplasm/immunology
- Tumor Microenvironment/immunology
- Animals
- Clinical Trials as Topic
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Safwaan H. Khan
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Yeonjoo Choi
- Division of Hematology/Oncology, Veterans Affairs (VA) Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Mysore Veena
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- Division of Hematology/Oncology, Veterans Affairs (VA) Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - John K. Lee
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Daniel Sanghoon Shin
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- Division of Hematology/Oncology, Veterans Affairs (VA) Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| |
Collapse
|
25
|
Shi J, Hou Z, Fan L, Hu C, Ma N, Huang E. Development and experimental validation of a senescence-related long non-coding RNA signature for prognostic prediction and immune microenvironment characterization in gastric cancer patients. J Gastrointest Oncol 2024; 15:2413-2436. [PMID: 39816024 PMCID: PMC11732332 DOI: 10.21037/jgo-24-792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 12/18/2024] [Indexed: 01/18/2025] Open
Abstract
Background Cellular senescence is considered a new marker of cancer. It has been suggested that long non-coding RNA (lncRNA) can be used to predict the prognosis of cancers. However, it remains to be seen whether the lncRNAs associated with cellular senescence can be used to predict the prognosis of gastric cancer (GC). The present study aimed to develop a novel senescence-related lncRNA signature (SenLncSig) to predict GC prognosis. The SenLncSig model holds promise for enhancing patient stratification, enabling more precise prognostic predictions and facilitating immunotherapy strategies. Methods Senescence-associated lncRNAs were identified from RNA expression profiles in The Cancer Genome Atlas (TCGA) database through the construction of a co-expression network linking senescence genes and lncRNAs. A prognostic signature for GC (334 patients from TCGA-STAD data set), comprising the senescence-related lncRNAs, was developed through univariate and multivariate Cox proportional hazards regression analyses. By using the median SenLncSig risk score, the GC patients were categorized into high- and low-risk groups. A Kaplan-Meier analysis and gene set enrichment analysis were conducted, and immune infiltration, the tumor mutation burden (TMB), and pharmacological treatments were compared between the high- and low-risk groups. We used an independent GC cohort (an external cohort of 30 pairs of tumor and non-tumor tissues from the GC patients) and three GC cell lines to conduct a quantitative reverse-transcription polymerase chain reaction (qRT-PCR) analysis to validate the results. Results We established a SenLncSig, a prognostic risk model comprising the following five senescence-associated lncRNAs; AP000695.2, LINC02381, AC005586.1, AP003392.1, and AP001528.2. According to the SenLncSig, high-risk scores were associated with poor overall survival (multivariate Cox proportional hazard ratio: 1.498, 95% confidence interval: 1.294-1.735; P<0.001). The time-dependent receiver operating characteristic curve indicated that the model performed (area under the curve: 0.711). We developed a nomogram incorporating age, gender, grade, stage, T stage, M stage, N stage, and SenLncSig risk score to estimate 1-year, 3-year, and 5-year survival rates. Further, according to the results of the mutation analysis, patients with a high TMB in the high-risk group had the worst prognosis. Interestingly, the high-risk group had a stronger infiltration of regulatory T cells (P<0.001) and M2 macrophage cells (P<0.001), as well as higher tumor immune dysfunction and exclusion scores than the low-risk group. These results might explain why the high-risk group had a worse prognosis. Finally, the qRT-PCR validation revealed that the AP000695.2 and AP003392.1 expression levels were significantly higher in the tumor tissues and GC cell lines than the normal tissues and normal human gastric epithelial cell line, whereas the opposite pattern was found for LINC02381. Conclusions The development of the SenLncSig provided a potential tool for improving patient prognosis predictions and offered preliminary insights into predicting the efficacy of GC immunotherapy.
Collapse
Affiliation(s)
- Jinglong Shi
- Department of General Surgery, Guangzhou Twelfth People’s Hospital, Guangzhou, China
| | - Zehui Hou
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ludi Fan
- Department of Guang Yuan Internal Medicine, Guangzhou Twelfth People’s Hospital, Guangzhou, China
| | - Chen Hu
- Department of General Surgery, Guangzhou Twelfth People’s Hospital, Guangzhou, China
| | - Ning Ma
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Enmin Huang
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
26
|
Sutton MN, Glazer SE, Al Zaki A, Napoli A, Yang P, Bhosale P, Liu J, Gammon ST, Piwnica-Worms D. Statins inhibit onco-dimerization of the 4Ig isoform of B7-H3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.18.628944. [PMID: 39763965 PMCID: PMC11702627 DOI: 10.1101/2024.12.18.628944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
B7-H3 (CD276), a member of the B7-family of immune checkpoint proteins, has been shown to have immunological and non-immunological effects promoting tumorigenesis [1, 2] and expression correlates with poor prognosis for many solid tumors, including cervical, ovarian and breast cancers [3-6]. We recently identified a tumor-cell autochthonous tumorigenic role for dimerization of the 4Ig isoform of B7-H3 (4Ig-B7-H3) [7], where 4Ig-B7-H3 dimerization in cis activated tumor-intrinsic cellular proliferation and tumorigenesis pathways, providing a novel opportunity for therapeutic intervention. Herein, a live cell split-luciferase complementation strategy was used to visualize 4Ig-B7-H3 homodimerization in a high-throughput small molecule screen (HTS) to identify modulators of this protein-protein interaction (PPI). Notably, the HTS identified several compounds that converged on lipid metabolism (including HMG-CoA reductase inhibitors, also known as statins) as significant inhibitors of 4Ig-B7-H3 dimerization (p < 0.01). In vitro and in vivo murine studies provided evidence that statin-mediated disruption of 4Ig-B7-H3 dimerization was associated with anti-tumor effects. Statin-mediated anti-cancer efficacy was selective for B7-H3-expressing tumors and retrospective analysis of clinical tumor specimens supported the hypothesis that concurrent statin use enhanced clinical outcomes for patients in a B7-H3 restricted manner. Thus, disruption of 4Ig-B7-H3 dimerization provides an unanticipated molecular mechanism linking statin use in cancer therapy and prevention with immune checkpoint.
Collapse
Affiliation(s)
- Margie N. Sutton
- Department of Cancer Systems Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Sarah E. Glazer
- Department of Cancer Systems Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ajlan Al Zaki
- Department of Cancer Systems Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Arianna Napoli
- Department of Cancer Systems Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ping Yang
- Department of Cancer Systems Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Priya Bhosale
- Department of Abdominal Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Jinsong Liu
- Department of Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Seth T. Gammon
- Department of Cancer Systems Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - David Piwnica-Worms
- Department of Cancer Systems Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
27
|
Zhang X, Zhang L, Cui M, Ji S, Zhang Y, Li Q, Zhang M. SPAG5 is a potential therapeutic target affecting proliferation, apoptosis, and invasion of esophageal cancer cells. Eur J Med Res 2024; 29:596. [PMID: 39696708 DOI: 10.1186/s40001-024-02182-y] [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: 10/17/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Sperm-associated antigen 5 (SPAG5) is a mitotic spindle protein crucial for coordinating the separation of sister chromatids into daughter cells. Increasing evidence suggests that SPAG5 is overexpressed in various malignancies, functioning as an oncogene. However, research specifically examining SPAG5 in esophageal cancer remains limited. METHODS In this research, we leveraged bioinformatics techniques to evaluate the expression and prognostic significance of SPAG5 in a variety of cancer types. We conducted Gene Set Enrichment Analysis (GSEA) to elucidate the relationship between SPAG5 and cancer characteristics. Additionally, we investigated the correlation between SPAG5 expression and immune cell infiltration utilizing the TIMER2.0 platform. The TIDE platform was used to assess the impact of SPAG5 on the effectiveness of immunotherapy and to screen for potential therapeutic drugs. We employed qRT-PCR and immunohistochemistry staining to ascertain the expression of SPAG5 in esophageal cancer tissue. Through cellular functional experiments, we examined the influence of SPAG5 expression on the proliferation, apoptosis, invasion, and migration of esophageal cancer cells. The Pathscan Stress Signaling Antibody Array was utilized to probe the potential molecular mechanisms of SPAG5. RESULTS SPAG5 exhibits high levels of expression in various cancers, encompassing esophageal cancer, and its presence indicates an unfavorable prognosis. SPAG5 is primarily enriched in pathways associated with cellular proliferation and demonstrates a correlation with immune gene expression as well as the infiltration of immune cells. Suppression of SPAG5 expression in esophageal cancer cells not only inhibits cell proliferation, but also attenuates cell invasion and migration while inducing cellular apoptosis. The depletion of SPAG5 results in a decline in the levels of critical signaling proteins. CONCLUSION SPAG5 plays a pivotal role in esophageal cancer cell proliferation, apoptosis, and metastasis within the tumor microenvironment, making it a promising therapeutic target.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, 710021, Shaanxi, China
| | - Lingmin Zhang
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Manli Cui
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, Shaanxi, China
- Engineering Research Center of Shaanxi Universities for Innovative Services of Chronic Disease Prevention and Control and Transformation of Nutritional Functional Food, Xi'an, 710077, Shaanxi, China
| | - Shiyu Ji
- Jingbian County People's Hospital of Shaanxi Province, Yulin Shi, 718500, Shaanxi, China
| | - Yanan Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, Shaanxi, China
| | - Qian Li
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, Shaanxi, China.
- Engineering Research Center of Shaanxi Universities for Innovative Services of Chronic Disease Prevention and Control and Transformation of Nutritional Functional Food, Xi'an, 710077, Shaanxi, China.
| | - Mingxin Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, Shaanxi, China.
- Engineering Research Center of Shaanxi Universities for Innovative Services of Chronic Disease Prevention and Control and Transformation of Nutritional Functional Food, Xi'an, 710077, Shaanxi, China.
| |
Collapse
|
28
|
Morita D, Rosewell Shaw A, Biegert G, Porter C, Woods M, Vasileiou S, Lim B, Suzuki M. Additional expression of T-cell engager in clinically tested oncolytic adeno-immunotherapy redirects tumor-infiltrated, irrelevant T cells against cancer cells to enhance antitumor immunity. J Immunother Cancer 2024; 12:e009741. [PMID: 39653552 PMCID: PMC11629014 DOI: 10.1136/jitc-2024-009741] [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: 05/28/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Oncolytic adenoviruses (OAds) are the most clinically tested viral vectors for solid tumors. However, most clinically tested "Armed" OAds show limited antitumor effects in patients with various solid tumors even with increased dosages and multiple injections. We developed a binary oncolytic/helper-dependent adenovirus system (CAdVEC), in which tumors are coinfected with an OAd and a non-replicating helper-dependent Ad (HDAd). We recently demonstrated that a single low-dose CAdVEC expressing interleukin-12, programmed death-ligand 1 blocker, and HSV thymidine kinase safety switch (CAdTrio) induces significant antitumor effects in patients, including complete response. Similar to previous OAd studies, all patients primarily amplified Ad-specific T cells after treatment however, CAdVEC was still able to induce clinical responses even given at a 100-fold lower dose. METHODS To address the mechanisms of CAdTrio-mediated antitumor effect in patients, we analyzed patients' samples using Enzyme-linked immunosorbent spot (ELISpot) to measure T-cell specificity and quantitative polymerase chain reaction (qPCR) to measure CAdVEC viral genome copies at tumor sites. We then evaluated potential mechanisms of CAdVEC efficacy in vitro using live-cell imaging. Based on those results, we developed a new CAdVEC additionally expressing a T-cell engager molecule targeting CD44v6 to redirect tumor-infiltrating irrelevant T cells against cancer stem cell populations (CAdTetra) for further improvement of local CAdVEC treatment. We tested its efficacy against different cancer types both in vitro and in vivo including Ad pre-immunized humanized mice. RESULTS We found that HDAd-infected cells escape Ad-specific T-cell recognition with enhanced tumor-specific T-cell activity through immunomodulatory transgenes. Since CAdVEC treatment initially amplified Ad-specific T cells in patients, we re-direct these virus-specific T cells to target tumor cells by additionally expressing CD44v6.BiTE from CAdTetra. CAdTetra significantly controlled tumor growth, repolarizing local and systemic responses against cancer cells in both immunologically "hot" and "cold" tumor models and also induced immunologic memory against rechallenged tumors. CONCLUSIONS Our results indicate that CAdTetra effectively induces adaptive T-cell responses against cancer cells by using tumor-infiltrating irrelevant T cells.
Collapse
Affiliation(s)
- Daisuke Morita
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX, USA
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan
| | - Amanda Rosewell Shaw
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX, USA
- Department of Biology, School of Science and Engineering, Benedict College, Columbia, SC, USA
| | - Greyson Biegert
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX, USA
| | - Caroline Porter
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX, USA
| | - Mae Woods
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX, USA
| | - Spyridoula Vasileiou
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Bora Lim
- Duncan Cancer Center-Breast, Baylor College of Medicine, Houston, TX, USA
- Breast Medical Oncology, The UT MD Anderson Cancer Center, Houston, TX, USA
| | - Masataka Suzuki
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
29
|
Gayen S, Mukherjee S, Dasgupta S, Roy S. Emerging druggable targets for immune checkpoint modulation in cancer immunotherapy: the iceberg lies beneath the surface. Apoptosis 2024; 29:1879-1913. [PMID: 39354213 DOI: 10.1007/s10495-024-02022-8] [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] [Accepted: 09/18/2024] [Indexed: 10/03/2024]
Abstract
The immune system serves as a fundamental defender against the initiation and progression of cancer. Failure of the immune system augments immunosuppressive action that leading to cancer manifestation. This immunosuppressive effect causes from significant alterations in immune checkpoint expression associated with tumoral progression. The tumor microenvironment promotes immune escape mechanisms that further amplifying immunosuppressive actions. Notably, substantial targeting of immune checkpoints has been pragmatic in the advancement of cancer research. This study highlights a comprehensive review of emerging druggable targets aimed at modulating immune checkpoint co-inhibitory as well as co-stimulatory molecules in response to immune system activation. This modulation has prompted to the development of newer therapeutic insights, eventually inducing immunogenic cell death through immunomodulatory actions. The study emphasizes the role of immune checkpoints in immunogenic regulation of cancer pathogenesis and explores potential therapeutic avenues in cancer immunotherapy.Modulation of Immunosuppressive and Immunostimulatory pathways of immune checkpoints in cancer immunotherapy.
Collapse
Affiliation(s)
- Sakuntala Gayen
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India
| | - Swarupananda Mukherjee
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India
| | - Sandipan Dasgupta
- Department of Pharmaceutical Technology, Maulana Abul Kalam Azad University of Technology, Kolkata, West Bengal, 741249, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India.
| |
Collapse
|
30
|
Liu L, Yao Z, Liu Y, Li Y, Ding Y, Hu J, Liu Z, Shi P, Chen K, Liu Z, Zhang W, Hou Y. A Pan-Cancer Analysis of the Oncogenic Role of CD276 in Human Tumors. Genes (Basel) 2024; 15:1527. [PMID: 39766794 PMCID: PMC11675885 DOI: 10.3390/genes15121527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
Objectives: B7 homolog 3 protein (B7-H3, also known as CD276) is a member of the B7 family that has been found to be associated with the growth and progression of a variety of tumors, but no pan-cancer evaluations of CD276 have been performed so far. In this study, we aimed to perform a pan-cancer analysis of the oncogenic role of CD276 in human tumors; Methods: We used a series of databases to perform a pan-cancer analysis of CD276, including the expression level of CD276 in pan-cancer and its relationship to tumor progression, patient survival duration, the immune cell infiltration within the tumor, and the potential signaling pathways and molecular mechanisms associated with CD276; Results: We found that CD276 was a potential biomarker for the prognosis of most cancers. The high expression of CD276 was associated with tumor progression, leading to poor survival. Notably, the up-regulation of CD276 expression in tumors increased the tumor infiltration of cancer-associated fibroblasts (CAFs) and myeloid-derived suppressor cells (MDSCs) and decreased the CD8+ T cells; Conclusions: Our study demonstrates that CD276 might promote tumor progression via the promotion of an immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Lilong Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhipeng Yao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yiting Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yang Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuhong Ding
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Junyi Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhenghao Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Pengjie Shi
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ke Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zheng Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wenhao Zhang
- Rehabilitation Medicine Center, The Affiliated Hospital of Hubei Provincial Government, Wuhan 430071, China;
| | - Yaxin Hou
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
31
|
Babič D, Jovčevska I, Zottel A. B7-H3 in glioblastoma and beyond: significance and therapeutic strategies. Front Immunol 2024; 15:1495283. [PMID: 39664380 PMCID: PMC11632391 DOI: 10.3389/fimmu.2024.1495283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/06/2024] [Indexed: 12/13/2024] Open
Abstract
Cancer has emerged as the second most prevalent disease and the leading cause of death, claiming the lives of 10 million individuals each year. The predominant varieties of cancer encompass breast, lung, colon, rectal, and prostate cancers. Among the more aggressive malignancies is glioblastoma, categorized as WHO stage 4 brain cancer. Following diagnosis, the typical life expectancy ranges from 12 to 15 months, as current established treatments like surgical intervention, radiotherapy, and chemotherapy using temozolomide exhibit limited effectiveness. Beyond conventional approaches, the exploration of immunotherapy for glioblastoma treatment is underway. A methodology involves CAR-T cells, monoclonal antibodies, ADCC and nanobodies sourced from camelids. Immunotherapy's recent focal point is the cellular ligand B7-H3, notably abundant in tumor cells while either scarce or absent in normal ones. Its expression elevates with cancer progression and serves as a promising prognostic marker. In this article, we delve into the essence of B7-H3, elucidating its function and involvement in signaling pathways. We delineate the receptors it binds to and its significance in glioblastoma and other cancer types. Lastly, we examine its role in immunotherapy and the utilization of nanobodies in this domain.
Collapse
|
32
|
Meeus F, Funeh CN, Awad RM, Zeven K, Autaers D, De Becker A, Van Riet I, Goyvaerts C, Tuyaerts S, Neyns B, Devoogdt N, De Vlaeminck Y, Breckpot K. Preclinical evaluation of antigen-sensitive B7-H3-targeting nanobody-based CAR-T cells in glioblastoma cautions for on-target, off-tumor toxicity. J Immunother Cancer 2024; 12:e009110. [PMID: 39562005 PMCID: PMC11575280 DOI: 10.1136/jitc-2024-009110] [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: 02/21/2024] [Accepted: 10/27/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Glioblastoma is the most common lethal primary brain tumor, urging evaluation of new treatment options. Chimeric antigen receptor (CAR)-T cells targeting B7 homolog 3 (B7-H3) are promising because of the overexpression of B7-H3 on glioblastoma cells but not on healthy brain tissue. Nanobody-based (nano)CARs are gaining increasing attention as promising alternatives to classical single-chain variable fragment-based (scFv)CARs, because of their single-domain nature and low immunogenicity. Still, B7-H3 nanoCAR-T cells have not been extensively studied in glioblastoma. METHODS B7-H3 nanoCAR- and scFvCAR-T cells were developed and evaluated in human glioblastoma models. NanoCAR-T cells targeting an irrelevant antigen served as control. T cell activation, cytokine secretion and killing capacity were evaluated in vitro using ELISA, live cell imaging and flow cytometry. Antigen-specific killing was assessed by generating B7-H3 knock-out cells using Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9-genome editing. The tumor tracing capacity of the B7-H3 nanobody was first evaluated in vivo using nuclear imaging. Then, the therapeutic potential of the nanoCAR-T cells was evaluated in a xenograft glioblastoma model. RESULTS We showed that B7-H3 nanoCAR-T cells were most efficient in lysing B7-H3pos glioblastoma cells in vitro. Lack of glioblastoma killing by control nanoCAR-T cells and lack of B7-H3neg glioblastoma killing by B7-H3 nanoCAR-T cells showed antigen-specificity. We showed in vivo tumor targeting capacity of the B7-H3 nanobody-used for the nanoCAR design-in nuclear imaging experiments. Evaluation of the nanoCAR-T cells in vivo showed tumor control in mice treated with B7-H3 nanoCAR-T cells in contrast to progressive disease in mice treated with control nanoCAR-T cells. However, we observed limiting toxicity in mice treated with B7-H3 nanoCAR-T cells and showed that the B7-H3 nanoCAR-T cells are activated even by low levels of mouse B7-H3 expression. CONCLUSIONS B7-H3 nanoCAR-T cells showed promise for glioblastoma therapy following in vitro characterization, but limiting in vivo toxicity was observed. Off-tumor recognition of healthy mouse tissue by the cross-reactive B7-H3 nanoCAR-T cells was identified as a potential cause for this toxicity, warranting caution when using highly sensitive nanoCAR-T cells, recognizing the low-level expression of B7-H3 on healthy tissue.
Collapse
Affiliation(s)
- Fien Meeus
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
- Department of Medical Imaging (MIMA), Molecular Imaging and Therapy (MITH) research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cyprine Neba Funeh
- Department of Medical Imaging (MIMA), Molecular Imaging and Therapy (MITH) research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Robin Maximilian Awad
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Katty Zeven
- Department of Medical Imaging (MIMA), Molecular Imaging and Therapy (MITH) research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Dorien Autaers
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Ann De Becker
- Department of Hematology, Cellular Therapy Laboratory, University Hospital Brussels, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Ivan Van Riet
- Department of Hematology, Cellular Therapy Laboratory, University Hospital Brussels, Universitair Ziekenhuis Brussel, Brussels, Belgium
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Hematology and Immunology Research Team (HEIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Cleo Goyvaerts
- Department of Medical Imaging (MIMA), Molecular Imaging and Therapy (MITH) research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sandra Tuyaerts
- Department of Medical Oncology, University Hospital Brussels, Universitair Ziekenhuis Brussel, Brussels, Belgium
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel, Brussels, Belgium
| | - Bart Neyns
- Department of Medical Oncology, University Hospital Brussels, Universitair Ziekenhuis Brussel, Brussels, Belgium
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel, Brussels, Belgium
| | - Nick Devoogdt
- Department of Medical Imaging (MIMA), Molecular Imaging and Therapy (MITH) research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yannick De Vlaeminck
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
33
|
Tsai KW, Liao JB, Tseng HW. Metformin regulates the proliferation and motility of melanoma cells by modulating the LINC00094/miR-1270 axis. Cancer Cell Int 2024; 24:384. [PMID: 39563323 PMCID: PMC11575040 DOI: 10.1186/s12935-024-03545-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Melanoma is an aggressive tumor with a high mortality rate. Metformin, a commonly prescribed diabetes medication, has shown promise in cancer prevention and treatment. Long noncoding RNAs (lncRNAs) are non-protein-coding RNA molecules that play a key role in tumor development by interacting with cellular chromatins. Despite the benefits of metformin, the anticancer mechanism underlying its effect on the regulation of lncRNAs in melanoma remains unclear. METHODS We investigated the lncRNA profiles of human melanoma cells with and without metformin treatment using a next-generation sequencing approach (NGS). Utilizing public databases, we analyzed the expression levels and clinical impacts of LINC00094 and miR-1270 in melanoma. The expression levels of LINC00094 and miR-1270 were verified in human cell lines and clinical samples by real-time PCR and in situ hybridization. The biological roles of LINC00094 and miR-1270 in cell growth, proliferation, cell cycle, apoptosis, and motility were studied using in vitro assays. RESULTS We identify a novel long noncoding RNA, namely LINC00094, whose expression considerably decreased in melanoma cells after metformin treatment. In situ hybridization analysis revealed substantially higher expression of LINC00094 in cutaneous melanoma tissue compared with adjacent normal epidermis and normal control tissues (P < 0.001). In nondiabetic patients with melanoma, the overall survival of high LINC00094 expression group was shorter than the low LINC00094 expression group with borderline statistical significance (log-rank test, P = 0.057). Coexpression analysis of LINC00094 indicated its involvement in the mitochondrial respiratory pathway, with its knockdown suppressing genes associated with mitochondrial oxidative phosphorylation, glycolysis, antioxidant production, and metabolite levels. Functional analysis revealed that silencing-LINC00094 inhibited the proliferation, colony formation, invasion, and migration of melanoma cells. Cell cycle analysis following LINC00094 knockdown revealed G1 phase arrest with reduced cell cycle protein expression. Combined TargetScan and reporter assays revealed a direct link between miR-1270 and LINC00094. Ectopic miR-1270 expression inhibited melanoma cell growth and motility while inducing apoptosis. Finally, through in silico analysis, we identified two miR-1270 target genes, CD276 and centromere protein M (CENPM), which may be involved in the biological functions of LINC00094. CONCLUSIONS Overall, LINC00094 expression may regulate melanoma cell growth and motility by modulating the expression of miR-1270, and targeting genes of CD276 and CENPM indicating its therapeutic potential in melanoma treatment.
Collapse
Affiliation(s)
- Kuo-Wang Tsai
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- Department of Nursing, Cardinal Tien Junior College of Healthcare and Management, New Taipei City, Taiwan
| | - Jia-Bin Liao
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Shu Zen Junior College of Medicine and Management, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Hui-Wen Tseng
- Department of Dermatology, Ministry of Health and Welfare Pingtung Hospital, Pingtung, Taiwan.
- Institute of Biomedical Sciences, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan.
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan.
- Department of Nursing, College of Nursing, Meiho University, Neipu, Pingtung, Taiwan.
| |
Collapse
|
34
|
Tigabu A. Immunoregulatory protein B7-H3 upregulated in bacterial and viral infection and its diagnostic potential in clinical settings. Front Immunol 2024; 15:1472626. [PMID: 39497833 PMCID: PMC11532155 DOI: 10.3389/fimmu.2024.1472626] [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: 07/29/2024] [Accepted: 09/30/2024] [Indexed: 11/07/2024] Open
Abstract
Bacterial and viral infections cause a huge burden to healthcare settings worldwide, and mortality rates associated with infectious microorganisms have remained high in recent decades. Despite tremendous efforts and resources worldwide to explore diagnostic biomarkers, rapid and easily assayed indicators for the diagnosis of bacterial and viral infections remain a challenge. B7 homolog 3 (B7-H3), a member of the B7 family of immunoregulatory proteins, is overexpressed in patients with septicemia, meningitis, pneumonia, and hepatitis. Therefore, B7-H3 could be used as a potential clinical indicator and therapeutic target for bacterial and viral infections caused by H. pylori, S. pneumoniae, M. pneumoniae, hepatitis B virus (HBV), viral hemorrhagic septicemia virus (VHSV), respiratory syncytial virus (RSV), and human immunodeficiency virus (HIV). Moreover, the interplay between infectious microorganisms and B7-H3 and exploration of the functional roles of the B7-H3 molecule could aid in the development of novel strategies for disease diagnosis and immunotherapy.
Collapse
Affiliation(s)
- Abiye Tigabu
- Department of Medical Microbiology, University of Gondar,
Gondar, Ethiopia
| |
Collapse
|
35
|
Cheng Y, Lin X, Xu H, Xu M, Xie W, Zeng J, Cui B, Cheng Z, Zhao J, Sun Y. Integrin β5, a noninvasive diagnostic biomarker, is associated with unfavorable prognosis and immunotherapy efficacy in gastric cancer. BMC Gastroenterol 2024; 24:362. [PMID: 39394072 PMCID: PMC11470723 DOI: 10.1186/s12876-024-03447-w] [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: 06/05/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Integrin β5 (ITGB5) is a pivotal player in the pathogenesis of gastric cancer (GC). We aimed to explore the potential value of ITGB5 as a predictor of diagnosis and immunotherapy in gastric cancer. METHODS The expression of ITGB5 in GC was assessed using The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, and verified through quantitative polymerase chain reaction (qPCR) and immunohistochemistry. Kaplan-Meier curves were conducted to evaluate the prognostic significance. The immune cells infiltration, tumor mutational burden (TMB), and immunophenoscore (IPS) were examined using CIBERSORT, TIMER, and TISIDB. In addition, colony formation, scratch assays, and transwell assays were employed to determine the impact on tumor progression and metastasis. CD276 expression was detected by western blotting following the knockdown of ITGB5. ELISA was utilized to measure serum ITGB5 levels. RESULTS The expression of ITGB5 in GC tissue surpassed that in normal tissue, it might contribute to GC pathogenesis through pathways including PI3K-AKT, ECM-receptor interaction, and TGF-beta. The elevated ITGB5 expression is associated with poor prognosis in GC patients. In addition, a strong positive association between ITGB5 overexpression and the infiltration levels of macrophages and monocytes, and it significantly influenced immune response. Moreover, lower expression of ITGB5 was associated with better immunotherapy efficacy. Subsequent investigation demonstrated that silencing of ITGB5 suppressed the proliferation and migration of GC cell lines in vitro. ITGB5 expression was positively correlated with CD276 expression and the knockdown of ITGB5 resulted a notable decrease CD276 expression. Futhermore, a significantly high level of serum ITGB5 was observed in GC patients. The combined assessment of ITGB5, CEA, and CA19-9 improved the diagnostic accuracy. CONCLUSIONS ITGB5 potentially serve as both a diagnostic biomarker and therapeutic target in managing GC.
Collapse
Affiliation(s)
- Yangyang Cheng
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xin Lin
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Huimin Xu
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Mingcheng Xu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Wanlin Xie
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Jingya Zeng
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Baohong Cui
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Zexu Cheng
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Jing Zhao
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Yihua Sun
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
36
|
Novysedlak R, Guney M, Al Khouri M, Bartolini R, Koumbas Foley L, Benesova I, Ozaniak A, Novak V, Vesely S, Pacas P, Buchler T, Ozaniak Strizova Z. The Immune Microenvironment in Prostate Cancer: A Comprehensive Review. Oncology 2024:1-25. [PMID: 39380471 DOI: 10.1159/000541881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 10/04/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND Prostate cancer (PCa) is a malignancy with significant immunosuppressive properties and limited immune activation. This immunosuppression is linked to reduced cytotoxic T cell activity, impaired antigen presentation, and elevated levels of immunosuppressive cytokines and immune checkpoint molecules. Studies demonstrate that cytotoxic CD8+ T cell infiltration correlates with improved survival, while increased regulatory T cells (Tregs) and tumor-associated macrophages (TAMs) are associated with worse outcomes and therapeutic resistance. Th1 cells are beneficial, whereas Th17 cells, producing interleukin-17 (IL-17), contribute to tumor progression. Tumor-associated neutrophils (TANs) and immune checkpoint molecules, such as PD-1/PD-L1 and T cell immunoglobulin-3 (TIM-3) are also linked to advanced stages of PCa. Chemotherapy holds promise in converting the "cold" tumor microenvironment (TME) to a "hot" one by depleting immunosuppressive cells and enhancing tumor immunogenicity. SUMMARY This comprehensive review examines the immune microenvironment in PCa, focusing on the intricate interactions between immune and tumor cells in the TME. It highlights how TAMs, Tregs, cytotoxic T cells, and other immune cell types contribute to tumor progression or suppression and how PCa's low immunogenicity complicates immunotherapy. KEY MESSAGES The infiltration of cytotoxic CD8+ T cells and Th1 cells correlates with better outcomes, while elevated T regs and TAMs promote tumor growth, metastasis, and resistance. TANs and natural killer (NK) cells exhibit dual roles, with higher NK cell levels linked to better prognoses. Immune checkpoint molecules like PD-1, PD-L1, and TIM-3 are associated with advanced disease. Chemotherapy can improve tumor immunogenicity by depleting T regs and myeloid-derived suppressor cells, offering therapeutic promise.
Collapse
Affiliation(s)
- Rene Novysedlak
- Third Department of Surgery, 1st Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Miray Guney
- Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Majd Al Khouri
- Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Robin Bartolini
- Lausanne Center for Immuno-oncology Toxicities (LCIT), Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Lily Koumbas Foley
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Iva Benesova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Andrej Ozaniak
- Third Department of Surgery, 1st Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Vojtech Novak
- Department of Urology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Stepan Vesely
- Department of Urology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Pavel Pacas
- Department of Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Tomas Buchler
- Department of Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Zuzana Ozaniak Strizova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| |
Collapse
|
37
|
Hagelstein I, Wessling L, Rochwarger A, Zekri L, Klimovich B, Tegeler CM, Jung G, Schürch CM, Salih HR, Lutz MS. Targeting CD276 for T cell-based immunotherapy of breast cancer. J Transl Med 2024; 22:902. [PMID: 39367484 PMCID: PMC11452943 DOI: 10.1186/s12967-024-05689-4] [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: 06/14/2024] [Accepted: 09/17/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND Breast cancer (BC) is the most common malignancy in women. Immunotherapy has revolutionized treatment options in many malignancies, and the introduction of immune checkpoint inhibition yielded beneficial results also in BC. However, many BC patients are ineligible for this T cell-based therapy, others do not respond or only briefly. Thus, there remains a high medical need for new therapies, particularly for triple-negative BC. CD276 (B7-H3) is overexpressed in several tumors on both tumor cells and tumor vessels, constituting a promising target for immunotherapy. METHODS We analyzed tumor samples of 25 patients using immunohistochemistry to assess CD276 levels. The potential of CC-3, a novel bispecific CD276xCD3 antibody, for BC treatment was evaluated using various functional in vitro assays. RESULTS Pronounced expression of CD276 was observed in all analyzed tumor samples including triple negative BC. In analyses with BC cells, CC-3 induced profound T cell activation, proliferation, and T cell memory subset formation. Moreover, treatment with CC-3 induced cytokine secretion and potent tumor cell lysis. CONCLUSION Our findings characterize CD276 as promising target and preclinically document the therapeutic potential of CC-3 for BC treatment, providing a strong rationale for evaluation of CC-3 in BC patients in a clinical trial for which the recruitment has recently started.
Collapse
Affiliation(s)
- Ilona Hagelstein
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, German Cancer Consortium (DKTK), University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Laura Wessling
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, German Cancer Consortium (DKTK), University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
| | - Alexander Rochwarger
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Latifa Zekri
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, German Cancer Consortium (DKTK), University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- Department for Immunology and German Cancer Consortium (DKTK), Eberhard Karls University, Tübingen, Germany
| | - Boris Klimovich
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, German Cancer Consortium (DKTK), University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Christian M Tegeler
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, German Cancer Consortium (DKTK), University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- Department of Obstetrics and Gynecology, University Hospital Tübingen, Tübingen, Germany
- Department of Peptide-Based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
| | - Gundram Jung
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- Department for Immunology and German Cancer Consortium (DKTK), Eberhard Karls University, Tübingen, Germany
| | - Christian M Schürch
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Helmut R Salih
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, German Cancer Consortium (DKTK), University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany.
| | - Martina S Lutz
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, German Cancer Consortium (DKTK), University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| |
Collapse
|
38
|
Ranganathan S, Reddy A, Russo A, Malepelle U, Desai A. Double agents in immunotherapy: Unmasking the role of antibody drug conjugates in immune checkpoint targeting. Crit Rev Oncol Hematol 2024; 202:104472. [PMID: 39111458 DOI: 10.1016/j.critrevonc.2024.104472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024] Open
Abstract
Antibody-drug conjugates (ADCs) have high specificity with lesser off-target effects, thus providing improved efficacy over traditional chemotherapies. A total of 14 ADCs have been approved for use against cancer by the US Food and Drug Administration (FDA), with more than 100 ADCs currently in clinical trials. Of particular interest ADCs targeting immune antigens PD-L1, B7-H3, B7-H4 and integrins. Specifically, we describe ADCs in development along with the gene and protein expression of these immune checkpoints across a wide range of cancer types let url = window.clickTag || window.clickTag1 || window.clickTag2 || window.clickTag3 || window.clickTag4 || window.bsClickTAG || window.bsClickTAG1 || window.bsClickTAG2 || window.url || ''; if(typeof url == 'string'){ document.body.dataset['perxceptAdRedirectUrl'] = url;}.
Collapse
Affiliation(s)
| | | | | | - Umberto Malepelle
- Department of Public Health University Federico II of Naples, Naples, Italy
| | - Aakash Desai
- Division of Hematology and Oncology, Department of Medicine, University of Alabama, Birmingham, United States.
| |
Collapse
|
39
|
Yu YH, Xu JH, Chen H, Lin YX, Ou-Yang J, Zhang ZY. CD276 is a promising biomarker for the prognosis of clear cell renal cell carcinoma. Kaohsiung J Med Sci 2024; 40:926-933. [PMID: 39210603 DOI: 10.1002/kjm2.12891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
This study aimed to investigate the role of cluster of differentiation 276 (CD276) in evaluating the prognosis of clear cell renal carcinoma (ccRCC) and to build a nomogram for predicting ccRCC progression post-surgery. Using data downloaded from The Cancer Genome Atlas (TCGA) database, we constructed a Kaplan-Meier (KM) curve depicting the relationship between CD276 expression levels and the progression-free interval (PFI) in 539 ccRCC cases. We further validated this by plotting a KM curve of the relationship between CD276 expression levels and PFI in 116 ccRCC patients from our hospital. Using clinical data collected from 116 patients, we identified independent risk factors affecting postoperative PFI in patients with ccRCC through univariate and multivariate COX analyses and created a nomogram for visual representation. Both TCGA and clinical data revealed a negative correlation between the expression levels of CD276 and PFI (p < 0.05). Univariate COX analysis revealed that the prognostic nutritional index, neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, systemic inflammatory index, World Health Organization grading, tumor diameter, CD276 expression levels, T stage, and N stage were related to PFI (p < 0.05). Furthermore, multivariate COX analysis indicated that tumor diameter and CD276 expression levels were independent risk factors for postoperative PFI in patients with ccRCC (p < 0.05). The calibration curve of the established nomogram exhibited a slope close to 1, with a Hosmer-Lemeshow goodness-of-fit test result of 2.335 and a p-value of 0.311. In patients with ccRCC, a negative correlation was noted between tumor CD276 expression and PFI. The larger the tumor diameter and the higher the tumor CD276 expression level, the shorter is the PFI.
Collapse
Affiliation(s)
- Yan-Hang Yu
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian-Hao Xu
- Department of Pathology, The First People's Hospital of Kunshan, Suzhou, China
| | - Hao Chen
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu-Xin Lin
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Ou-Yang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhi-Yu Zhang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
40
|
Zhou Q, Xu J, Chen X, Ouyang J, Mao C, Zhang Z. CD276 as a promising diagnostic and prognostic biomarker for bladder cancer through bioinformatics and clinical research. Front Oncol 2024; 14:1445526. [PMID: 39319055 PMCID: PMC11419956 DOI: 10.3389/fonc.2024.1445526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/19/2024] [Indexed: 09/26/2024] Open
Abstract
Objective To assess CD276 expression and explore its relationship with the clinicopathological characteristics and prognosis of patients with bladder cancer. Methods In total, RNA-sequencing data and clinical profiles of 436 bladder cancer cases from The Cancer Genome Atlas (TCGA) were assessed using the University of California Santa Cruz Xena (UCSC) platform. We compared the CD276 levels in cancerous and adjacent normal tissues and used the R software for statistical association with the clinical stage, grade, and survival (the overall survival, disease-specific survival, and progression-free survival). A single-gene GSEA analysis on TCGA-BLCA data was performed to explore potential pathways through which CD276 might influence bladder cancer. Additionally, CD276 expression was analyzed by comparing data from 9 cancerous tissues and 3 adjacent normal tissues in the GEO dataset GSE7476. Furthermore, we analyzed 133 cancerous bladder and adjacent tissue samples from the Soochow University Hospital, collected between January 1, 2016, and September 30, 2022, to assess the CD276 protein expression using immunohistochemistry. We examined the relationship between tumor CD276 levels and clinical outcomes and prognosis of bladder cancer. Results Bioinformatic analysis revealed elevated CD276 expression in tumors compared to that in adjacent tissues (p<0.05), correlating with poor survival. GSEA revealed that CD276 was significantly involved in extracellular matrix-related pathways. Immunohistochemistry confirmed CD276 overexpression in tumor tissues, with higher levels linked to advanced pathological grades and worse prognosis. Conclusion CD276 is markedly upregulated in bladder cancer and associated with severe pathological features, advanced disease, potential for metastasis, and diminished survival rates. It may promote bladder cancer development and progression by influencing extracellular matrix-related-related pathways, making it a viable diagnostic and prognostic biomarker for bladder cancer.
Collapse
Affiliation(s)
- Qi Zhou
- Department of Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianhao Xu
- Department of Pathology, The First People’s Hospital of Kunshan, Suzhou, China
| | - Xuelei Chen
- Department of Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Ouyang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Caiping Mao
- Department of Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhiyu Zhang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
41
|
Zhu X, Hu C, Zhang Z, Zhu Y, Liu W, Zheng B, Feng X, Lu H. PD-L1 and B7-H3 are Effective Prognostic Factors and Potential Therapeutic Targets for High-Risk Thyroid Cancer. Endocr Pathol 2024; 35:230-244. [PMID: 39102163 DOI: 10.1007/s12022-024-09822-3] [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] [Accepted: 07/24/2024] [Indexed: 08/06/2024]
Abstract
The prognosis of thyroid cancer in patients varies significantly based on different pathological types or distinct clinical situations. Investigating the expression of immune checkpoint molecules PD-L1 and B7-H3 in high-risk thyroid cancer and their correlation with clinicopathological features and prognosis will contribute to the development of novel therapeutic strategies. A retrospective sample of 202 patients with thyroid cancer who underwent surgery at the Cancer Hospital of the Chinese Academy of Medical Sciences was collected, including 33 cases of anaplastic thyroid cancer (ATC), 21 cases of differentiated thyroid cancer (DTC) with distant metastasis (DM), 7 cases of differentiated high-grade thyroid carcinoma (DHGTC), and 109 cases of aggressive subtypes of papillary thyroid carcinoma (PTC) (including 28 cases of tall cell PTC, 31 cases of diffuse sclerosing PTC, 20 cases of solid PTC, 15 cases of columnar cell PTC, and 15 cases of hobnail PTC). In the control group, there were 32 cases of classic PTC. The differences in protein expression between PD-L1 and B7-H3 in several high-risk thyroid cancers and normal tissues and controls were compared by immunohistochemical staining, and the clinicopathological features and prognostic relevance were statistically analyzed. The expression of PD-L1 in ATC (P < 0.001), tall cell PTC (P = 0.031), and DHGTC (P = 0.003) was significantly higher than that in classic PTC. The expression of B7-H3 in ATC (P < 0.001), DTC with DM (P = 0.001), diffuse sclerosing PTC (P = 0.013), columnar cell PTC (P = 0.007), solid PTC (P < 0.001), hobnail PTC (P < 0.001), and DHGTC (P < 0.001) was significantly higher than that in classic PTC. In ATC, PD-L1 expression correlated significantly with extrathyroidal extension (ETE) (P = 0.027) and B7-H3 expression correlated significantly with male patients (P = 0.031) and lymph node metastasis (LNM) (P = 0.026). The positive expression of B7-H3 (P = 0.041) was an independent risk factor for disease progression in ATC. B7-H3 positive expression (P = 0.049), PD-L1 positive expression (P = 0.015), and tumor diameter ≥ 2 cm (P = 0.038) were independent risk factors for disease progression in patients with DTC with DM. PD-L1 positive expression (P = 0.019) and tumor diameter ≥ 2 cm (P = 0.018) were independent risk factors for disease progression in patients with aggressive subtypes of PTC. B7-H3 and PD-L1 are expected to be effective prognostic indicators for patients with aggressive thyroid cancer, which can help in optimization of individualized treatment strategies. Immunotherapy targeting these two molecules may provide new and complementary ideas for the treatment of high-risk/refractory thyroid cancer.
Collapse
Affiliation(s)
- Xinyi Zhu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17# Panjiayuan Street, Chaoyang District, Beijing, 100021, China
| | - Chunfang Hu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17# Panjiayuan Street, Chaoyang District, Beijing, 100021, China
| | - Zhe Zhang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17# Panjiayuan Street, Chaoyang District, Beijing, 100021, China
| | - Yuelu Zhu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17# Panjiayuan Street, Chaoyang District, Beijing, 100021, China
| | - Wenchao Liu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17# Panjiayuan Street, Chaoyang District, Beijing, 100021, China
| | - Bo Zheng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17# Panjiayuan Street, Chaoyang District, Beijing, 100021, China
| | - Xiaoli Feng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17# Panjiayuan Street, Chaoyang District, Beijing, 100021, China.
| | - Haizhen Lu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17# Panjiayuan Street, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
42
|
He A, Huang Z, Feng Q, Zhang S, Li F, Li D, Lu H, Wang J. AC099850.3 promotes HBV-HCC cell proliferation and invasion through regulating CD276: a novel strategy for sorafenib and immune checkpoint combination therapy. J Transl Med 2024; 22:809. [PMID: 39217342 PMCID: PMC11366154 DOI: 10.1186/s12967-024-05576-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/04/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND This study investigates the molecular mechanisms of CC@AC&SF@PP NPs loaded with AC099850.3 siRNA and sorafenib (SF) for improving hepatitis B virus-related hepatocellular carcinoma (HBV-HCC). METHODS A dataset of 44 HBV-HCC patients and their survival information was selected from the TCGA database. Immune genes related to survival status were identified using the ImmPort database and WGCNA analysis. A prognostic risk model was constructed and analyzed using Lasso regression. Differential analysis was performed to screen key genes, and their significance and predictive accuracy for HBV-HCC were validated using Kaplan-Meier survival curves, ROC analysis, CIBERSORT analysis, and correlation analysis. The correlation between AC099850.3 and the gene expression matrix was calculated, followed by GO and KEGG enrichment analysis using AC099850.3 and its co-expressed genes. HepG2.2.15 cells were selected for in vitro validation, and lentivirus interference, cell cycle determination, CCK-8 experiments, colony formation assays, Transwell experiments, scratch experiments, and flow cytometry were performed to investigate the effects of key genes on HepG2.2.15 cells. A subcutaneous transplanted tumor model in mice was constructed to verify the inhibitory effect of key genes on HBV-HCC tumors. Subsequently, pH-triggered drug release NPs (CC@AC&SF@PP) were prepared, and their therapeutic effects on HBV-HCC in situ tumor mice were studied. RESULTS A prognostic risk model (AC012313.9, MIR210HG, AC099850.3, AL645933.2, C6orf223, GDF10) was constructed through bioinformatics analysis, showing good sensitivity and specificity in diagnostic prediction. AC099850.3 was identified as a key gene, and enrichment analysis revealed its impact on cell cycle pathways. In vitro cell experiments demonstrated that AC099850.3 promotes HepG2.2.15 cell proliferation and invasion by regulating immune checkpoint CD276 expression and cell cycle progression. In vivo, subcutaneously transplanted tumor experiments showed that AC099850.3 promotes the growth of HBV-HCC tumors in nude mice. Furthermore, pH-triggered drug release NPs (CC@AC&SF@PP) loaded with AC099850.3 siRNA and SF were successfully prepared and delivered to the in situ HBV-HCC, enhancing the effectiveness of combined therapy for HBV-HCC. CONCLUSIONS AC099850.3 accelerates the cell cycle progression and promotes the occurrence and development of HBV-HCC by upregulating immune checkpoint CD276 expression. CC@AC&SF@PP NPs loaded with AC099850.3 siRNA and SF improve the effectiveness of combined therapy for HBV-HCC.
Collapse
Affiliation(s)
- Aoxiao He
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, 330006, China
| | - Zhihao Huang
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, 330006, China
| | - Qian Feng
- Department of Emergency, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Shan Zhang
- Department of Hematology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Fan Li
- Department of Gastroenterology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Dan Li
- Department of Gastroenterology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Hongcheng Lu
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, 330006, China.
| | - Jiakun Wang
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, 330006, China.
| |
Collapse
|
43
|
Perovic D, Dusanovic Pjevic M, Perovic V, Grk M, Rasic M, Milickovic M, Mijovic T, Rasic P. B7 homolog 3 in pancreatic cancer. World J Gastroenterol 2024; 30:3654-3667. [PMID: 39193002 PMCID: PMC11346158 DOI: 10.3748/wjg.v30.i31.3654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/24/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024] Open
Abstract
Despite advances in cancer treatment, pancreatic cancer (PC) remains a disease with high mortality rates and poor survival outcomes. The B7 homolog 3 (B7-H3) checkpoint molecule is overexpressed among many malignant tumors, including PC, with low or absent expression in healthy tissues. By modulating various immunological and nonimmunological molecular mechanisms, B7-H3 may influence the progression of PC. However, the impact of B7-H3 on the survival of patients with PC remains a subject of debate. Still, most available scientific data recognize this molecule as a suppressive factor to antitumor immunity in PC. Furthermore, it has been demonstrated that B7-H3 stimulates the migration, invasion, and metastasis of PC cells, and enhances resistance to chemotherapy. In preclinical models of PC, B7-H3-targeting monoclonal antibodies have exerted profound antitumor effects by increasing natural killer cell-mediated antibody-dependent cellular cytotoxicity and delivering radioisotopes and cytotoxic drugs to the tumor site. Finally, PC treatment with B7-H3-targeting antibody-drug conjugates and chimeric antigen receptor T cells is being tested in clinical studies. This review provides a comprehensive analysis of all PC-related studies in the context of B7-H3 and points to deficiencies in the current data that should be overcome by future research.
Collapse
Affiliation(s)
- Dijana Perovic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Marija Dusanovic Pjevic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Vladimir Perovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Milka Grk
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Milica Rasic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Maja Milickovic
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia “Dr. Vukan Cupic”, Belgrade 11000, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Tanja Mijovic
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia “Dr. Vukan Cupic”, Belgrade 11000, Serbia
| | - Petar Rasic
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia “Dr. Vukan Cupic”, Belgrade 11000, Serbia
| |
Collapse
|
44
|
Park R, Yu J, Shahzad M, Lee S, Ji JD. The immune regulatory function of B7-H3 in malignancy: spotlight on the IFN-STAT1 axis and regulation of tumor-associated macrophages. Immunol Res 2024; 72:526-537. [PMID: 38265550 DOI: 10.1007/s12026-024-09458-9] [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: 11/29/2023] [Accepted: 01/17/2024] [Indexed: 01/25/2024]
Abstract
B7-H3 is a member of the B7 superfamily and a putative inhibitory immune checkpoint molecule. Several early-phase clinical trials have reported promising anti-tumor activity and safety of anti-cancer drugs targeting B7-H3, suggesting that it may be a promising target for a potential next-generation immune checkpoint inhibitor. Despite ongoing clinical studies, most B7-H3-targeted drugs being currently investigated rely on direct cytotoxicity as their mechanisms of action rather than modulating its function as an immune checkpoint, at least in part due to its incompletely understood immune regulatory function. Recent studies have begun to elucidate the role of B7-H3 in regulating the tumor microenvironment (TME). Emerging evidence suggests that B7-H3 may regulate the interferon-STAT1 axis in the TME and promote immune suppression. Similarly, increasing evidence shows B7-H3 may be implicated in promoting M1 to M2 polarization of tumor-associated macrophages (TAMs). There is also accumulating evidence suggesting that B7-H3 may play a role in the heterotypic fusion of cancer stem cells and macrophages, thereby promoting tumor invasion and metastasis. Here, we review the recent advances in the understanding of B7-H3 cancer immunobiology with a focus on highlighting its potential role in the interferon priming of TAMs and the heterotypic fusion of TAMs with cancer stem cells and suggest future direction in elucidating its immune checkpoint function.
Collapse
Affiliation(s)
- Robin Park
- Department of Hematology/Oncology, Moffitt Cancer Center/University of South Florida, Tampa, FL, USA
| | - James Yu
- Department of Hematology/Oncology, Moffitt Cancer Center/University of South Florida, Tampa, FL, USA
| | - Moazzam Shahzad
- Department of Hematology/Oncology, Moffitt Cancer Center/University of South Florida, Tampa, FL, USA
| | - Sunggon Lee
- Department of Internal Medicine, Korea University, Seoul, South Korea
| | - Jong Dae Ji
- Department of Rheumatology, College of Medicine, Korea University, Seoul, South Korea.
| |
Collapse
|
45
|
Meng Y, Wang X, Yang J, Zhu M, Yu M, Li L, Liang Y, Kong F. Antibody-drug conjugates treatment of small cell lung cancer: advances in clinical research. Discov Oncol 2024; 15:327. [PMID: 39090431 PMCID: PMC11294301 DOI: 10.1007/s12672-024-01171-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 07/16/2024] [Indexed: 08/04/2024] Open
Abstract
Small cell lung cancer (SCLC) is an extremely aggressive cancer with a relatively low median survival rate after diagnosis. Treatment options such as chemotherapy or combination immunotherapy have shown clinical benefits, but resistance and relapse can occur. Antibody-drug conjugates (ADCs), as a novel class of biopharmaceutical compounds, have broad application prospects in the treatment of SCLC. ADCs consist of monoclonal antibodies that specifically target cancer cells and are attached to cytotoxic drugs, allowing for targeted killing of cancer cells while sparing healthy tissues. Current clinical studies focus on Delta-like protein 3 (DLL3), CD56, Trophoblast cell surface antigen 2 (Trop-2), B7-H3, and SEZ6. Although toxicities exceeding expectations have been observed with Rova-T, drugs targeting TROP-2 (Sacituzumab Govitecan), B7-H3 (DS-7300), and SEZ6 (ABBV-011) have shown exciting clinical benefits. In this review, we collect the latest clinical evidence to describe the therapeutic efficacy and safety of ADCs in SCLC and discuss prospects and challenges.
Collapse
Affiliation(s)
- Yuan Meng
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin Cancer Institute of Traditional Chinese Medicine, Tianjin, China
| | - Xuerui Wang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin Cancer Institute of Traditional Chinese Medicine, Tianjin, China
| | - Jie Yang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin Cancer Institute of Traditional Chinese Medicine, Tianjin, China
| | - Meiying Zhu
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin Cancer Institute of Traditional Chinese Medicine, Tianjin, China
| | - Minghui Yu
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin Cancer Institute of Traditional Chinese Medicine, Tianjin, China
| | - Longhui Li
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin Cancer Institute of Traditional Chinese Medicine, Tianjin, China
| | - Yangyueying Liang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin Cancer Institute of Traditional Chinese Medicine, Tianjin, China
| | - Fanming Kong
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin, 300193, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
- Tianjin Cancer Institute of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
46
|
Deng T, Deng Y, Tsao ST, Xiong Q, Yao Y, Liu C, Gu MY, Huang F, Wang H. Rapidly-manufactured CD276 CAR-T cells exhibit enhanced persistence and efficacy in pancreatic cancer. J Transl Med 2024; 22:633. [PMID: 38978106 PMCID: PMC11229349 DOI: 10.1186/s12967-024-05462-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND Pancreatic cancer is one of the most lethal malignancies and the lack of treatment options makes it more deadly. Chimeric Antigen Receptor T-cell (CAR-T) immunotherapy has revolutionized cancer treatment and made great breakthroughs in treating hematological malignancies, however its success in treating solid cancers remains limited mainly due to the lack of tumor-specific antigens. On the other hand, the prolonged traditional manufacturing process poses challenges, taking 2 to 6 weeks and impacting patient outcomes. CD276 has recently emerged as a potential therapeutic target for anti-solid cancer therapy. Here, we investigated the efficacy of CD276 CAR-T and rapidly-manufactured CAR-T against pancreatic cancer. METHODS In the present study, CD276 CAR-T was prepared by CAR structure carrying 376.96 scFv sequence, CD8 hinge and transmembrane domain, 4-1BB and CD3ζ intracellular domains. Additionally, CD276 rapidly-manufactured CAR-T (named CD276 Dash CAR-T) was innovatively developed by shortening the duration of ex vitro culture to reduce CAR-T manufacturing time. We evaluated the anti-tumor efficacy of CD276 CAR-T and further compared the functional assessment of Dash CAR-T and conventional CAR-T in vitro and in vivo by detecting the immunophenotypes, killing ability, expansion capacity and tumor-eradicating effect of CAR-T. RESULTS We found that CD276 was strongly expressed in multiple solid cancer cell lines and that CD276 CAR-T could efficiently kill these solid cancer cells. Moreover, Dash CAR-T was successfully manufactured within 48-72 h and the functional validation was carried out subsequently. In vitro, CD276 Dash CAR-T possessed a less-differentiated phenotype and robust proliferative ability compared to conventional CAR-T. In vivo xenograft mouse model, CD276 Dash CAR-T showed enhanced anti-pancreatic cancer efficacy and T cell expansion. Besides, except for the high-dose group, the body weight of mice was maintained stable, and the state of mice was normal. CONCLUSIONS In this study, we proved CD276 CAR-T exhibited powerful activity against pancreatic cancer cells in vitro and in vivo. More importantly, we demonstrated the manufacturing feasibility, acceptable safety and superior anti-tumor efficacy of CD276 Dash CAR-T generated with reduced time. The results of the above studies indicated that CD276 Dash CAR-T immunotherapy might be a novel and promising strategy for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Tian Deng
- Department of Research and Development, Hrain Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong New District, Shanghai, China
| | - Yingzhi Deng
- Department of Research and Development, Hrain Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong New District, Shanghai, China
| | - Shih-Ting Tsao
- Department of Research and Development, Hrain Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong New District, Shanghai, China
| | - Qinghui Xiong
- Department of Research and Development, Hrain Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong New District, Shanghai, China
| | - Yue Yao
- Department of Research and Development, Hrain Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong New District, Shanghai, China
| | - Cuicui Liu
- Regulatory Affairs Department, Hrain Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong New District, Shanghai, China
| | - Ming Yuan Gu
- Department of Research and Development, Hrain Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong New District, Shanghai, China
| | - Fei Huang
- Department of Research and Development, Hrain Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong New District, Shanghai, China.
| | - Haiying Wang
- Department of Research and Development, Hrain Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong New District, Shanghai, China.
| |
Collapse
|
47
|
Wang Y, Lih TM, Lee JW, Ohtsuka T, Hozaka Y, Mino-Kenudson M, Adsay NV, Luchini C, Scarpa A, Maker AV, Kim GE, Paulino J, Chen L, Jiao L, Sun Z, Goodman D, Pflüger MJ, Roberts NJ, Matthaei H, Wood LD, Furukawa T, Zhang H, Hruban RH. Multi-omic profiling of intraductal papillary neoplasms of the pancreas reveals distinct expression patterns and potential markers of progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.07.602385. [PMID: 39005476 PMCID: PMC11245086 DOI: 10.1101/2024.07.07.602385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
In order to advance our understanding of precancers in the pancreas, 69 pancreatic intraductal papillary neoplasms (IPNs), including 64 intraductal papillary mucinous neoplasms (IPMNs) and 5 intraductal oncocytic papillary neoplasms (IOPNs), 32 pancreatic cyst fluid samples, 104 invasive pancreatic ductal adenocarcinomas (PDACs), 43 normal adjacent tissues (NATs), and 76 macro-dissected normal pancreatic ducts (NDs) were analyzed by mass spectrometry. A total of 10,246 proteins and 22,284 glycopeptides were identified in all tissue samples, and 756 proteins with more than 1.5-fold increase in abundance in IPMNs relative to NDs were identified, 45% of which were also identified in cyst fluids. The over-expression of selected proteins was validated by immunolabeling. Proteins and glycoproteins overexpressed in IPMNs included those involved in glycan biosynthesis and the immune system. In addition, multiomics clustering identified two subtypes of IPMNs. This study provides a foundation for understanding tumor progression and targets for earlier detection and therapies. Significance This multilevel characterization of intraductal papillary neoplasms of the pancreas provides a foundation for understanding the changes in protein and glycoprotein expression during the progression from normal duct to intraductal papillary neoplasm, and to invasive pancreatic carcinoma, providing a foundation for informed approaches to earlier detection and treatment.
Collapse
|
48
|
Qin H, Liu J, Qu Y, Li YY, Xu YL, Yan YF. The intratumoral microbiota biomarkers for predicting survival and efficacy of immunotherapy in patients with ovarian serous cystadenocarcinoma. J Ovarian Res 2024; 17:140. [PMID: 38970121 PMCID: PMC11227176 DOI: 10.1186/s13048-024-01464-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/26/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Ovarian serous cystadenocarcinoma, accounting for about 90% of ovarian cancers, is frequently diagnosed at advanced stages, leading to suboptimal treatment outcomes. Given the malignant nature of the disease, effective biomarkers for accurate prediction and personalized treatment remain an urgent clinical need. METHODS In this study, we analyzed the microbial contents of 453 ovarian serous cystadenocarcinoma and 68 adjacent non-cancerous samples. A univariate Cox regression model was used to identify microorganisms significantly associated with survival and a prognostic risk score model constructed using LASSO Cox regression analysis. Patients were subsequently categorized into high-risk and low-risk groups based on their risk scores. RESULTS Survival analysis revealed that patients in the low-risk group had a higher overall survival rate. A nomogram was constructed for easy visualization of the prognostic model. Analysis of immune cell infiltration and immune checkpoint gene expression in both groups showed that both parameters were positively correlated with the risk level, indicating an increased immune response in higher risk groups. CONCLUSION Our findings suggest that microbial profiles in ovarian serous cystadenocarcinoma may serve as viable clinical prognostic indicators. This study provides novel insights into the potential impact of intratumoral microbial communities on disease prognosis and opens avenues for future therapeutic interventions targeting these microorganisms.
Collapse
Affiliation(s)
- Hao Qin
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| | - Jie Liu
- Department of Medical Records, Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Yi Qu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 North Huayuan Road, Haidian District, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology, (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yang-Yang Li
- Medical Center for Human Reproduction, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ya-Lan Xu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, China
| | - Yi-Fang Yan
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 North Huayuan Road, Haidian District, Beijing, 100191, China.
- National Clinical Research Center for Obstetrics and Gynecology, (Peking University Third Hospital), Beijing, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
| |
Collapse
|
49
|
Stefańczyk SA, Hayn C, Heitmann J, Jung S, Zekri L, Märklin M. Expression and Prognostic Value of a Novel B7-H3 (CD276) Antibody in Acute Myeloid Leukemia. Cancers (Basel) 2024; 16:2455. [PMID: 39001517 PMCID: PMC11240323 DOI: 10.3390/cancers16132455] [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: 06/05/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Despite recent advances in immunophenotyping, the prognosis of acute myeloid leukemia (AML) is still mainly estimated using age and genetic markers. As the genetic heterogeneity of AML patients is high, flow cytometry-based classification with appropriate biomarkers can efficiently complement risk stratification and treatment selection. An increased expression of B7-H3 (CD276), an immune checkpoint protein, has been reported and associated with poor prognosis. However, the available data are limited and heterogeneous. Here, we used a novel, proprietary murine anti-B7-H3 8H8 antibody for the flow cytometric analysis of B7-H3 expression in AML blasts from 77 patients. Our antibody reliably detected substantial B7-H3 expression in 62.3% of AML patients. B7-H3 expression was higher in the monocytic French-American-British (FAB) M5 group and in intermediate and poor risk patients according to the European Leukemia Network. Using receiver operating characteristics (ROCs), we identified a specific fluorescence intensity cut-off of 4.45 to discriminate between B7-H3high and B7-H3low expression. High B7-H3 expression was associated with shorter overall survival (OS) and progression-free survival (PFS). In conclusion, we have developed a novel B7-H3 antibody that serves as a new tool for the detection of B7-H3 expression in AML and may help to facilitate risk stratification and treatment selection in AML patients.
Collapse
Affiliation(s)
- Sylwia A. Stefańczyk
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (S.A.S.); (C.H.); (J.H.); (S.J.)
- Cluster of Excellence iFIT (EXC 2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Clara Hayn
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (S.A.S.); (C.H.); (J.H.); (S.J.)
- Cluster of Excellence iFIT (EXC 2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Jonas Heitmann
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (S.A.S.); (C.H.); (J.H.); (S.J.)
- Cluster of Excellence iFIT (EXC 2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Susanne Jung
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (S.A.S.); (C.H.); (J.H.); (S.J.)
- Cluster of Excellence iFIT (EXC 2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
- Department of Peptide−based Immunotherapy, Institute of Immunology, University and University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Latifa Zekri
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (S.A.S.); (C.H.); (J.H.); (S.J.)
- Cluster of Excellence iFIT (EXC 2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Melanie Märklin
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (S.A.S.); (C.H.); (J.H.); (S.J.)
- Cluster of Excellence iFIT (EXC 2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| |
Collapse
|
50
|
Ren M, Zhang J, Zong R, Sun H. A Novel Pancreatic Cancer Hypoxia Status Related Gene Signature for Prognosis and Therapeutic Responses. Mol Biotechnol 2024; 66:1684-1703. [PMID: 37405638 DOI: 10.1007/s12033-023-00807-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 06/26/2023] [Indexed: 07/06/2023]
Abstract
Pancreatic cancer (PAC) is a highly fatal and aggressive type of cancer. Hypoxia is a common feature of PAC. The aim of this study was to develop a hypoxia status-related prognostic model for predicting the survival outcomes in PAC. The data sets of PAC from The Cancer Genome Atlas and the International Cancer Genome Consortium were used to construct and validate the signature. A 6 hypoxia status-related differential expression genes prognostic model for predicting the survival outcomes was established. The Kaplan-Meier analysis and Received operating characteristic curve indicated the good performance of the signature at predicting overall survival. Univariate and Multivariate Cox regression revealed that the signature was an independent prognostic factor in PAC. Weighted Gene Co-expression Network Analysis and immune infiltration analysis indicated that Immune-related pathways and immune cell infiltration was mostly enriched in the low-risk group, which presented a better prognosis. We also evaluated the predictive of the signature for immunotherapy and chemoradiotherapy. Risk gene LY6D may be a potential prognostic predictor of PAC. This model can be used as an independent prognostic factor for predicting clinical outcomes and a possible classifier for response to chemotherapy.
Collapse
Affiliation(s)
- Min Ren
- College of Life Science, Yan'an University, Yan'an, 716000, China.
| | - Jianing Zhang
- College of Life Science, Yan'an University, Yan'an, 716000, China
| | - Rongrong Zong
- College of Life Science, Yan'an University, Yan'an, 716000, China
| | - Huiru Sun
- College of Life Science, Yan'an University, Yan'an, 716000, China.
| |
Collapse
|