1
|
Vidman S, Ma YHE, Fullenkamp N, Plant GW. Human induced pluripotent stem cell-derived therapies for regeneration after central nervous system injury. Neural Regen Res 2025; 20:3063-3075. [PMID: 39715081 PMCID: PMC11881715 DOI: 10.4103/nrr.nrr-d-24-00901] [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: 08/09/2024] [Revised: 09/26/2024] [Accepted: 10/29/2024] [Indexed: 12/25/2024] Open
Abstract
In recent years, the progression of stem cell therapies has shown great promise in advancing the nascent field of regenerative medicine. Considering the non-regenerative nature of the mature central nervous system, the concept that "blank" cells could be reprogrammed and functionally integrated into host neural networks remained intriguing. Previous work has also demonstrated the ability of such cells to stimulate intrinsic growth programs in post-mitotic cells, such as neurons. While embryonic stem cells demonstrated great potential in treating central nervous system pathologies, ethical and technical concerns remained. These barriers, along with the clear necessity for this type of treatment, ultimately prompted the advent of induced pluripotent stem cells. The advantage of pluripotent cells in central nervous system regeneration is multifaceted, permitting differentiation into neural stem cells, neural progenitor cells, glia, and various neuronal subpopulations. The precise spatiotemporal application of extrinsic growth factors in vitro, in addition to microenvironmental signaling in vivo, influences the efficiency of this directed differentiation. While the pluri- or multipotency of these cells is appealing, it also poses the risk of unregulated differentiation and teratoma formation. Cells of the neuroectodermal lineage, such as neuronal subpopulations and glia, have been explored with varying degrees of success. Although the risk of cancer or teratoma formation is greatly reduced, each subpopulation varies in effectiveness and is influenced by a myriad of factors, such as the timing of the transplant, pathology type, and the ratio of accompanying progenitor cells. Furthermore, successful transplantation requires innovative approaches to develop delivery vectors that can mitigate cell death and support integration. Lastly, host immune responses to allogeneic grafts must be thoroughly characterized and further developed to reduce the need for immunosuppression. Translation to a clinical setting will involve careful consideration when assessing both physiologic and functional outcomes. This review will highlight both successes and challenges faced when using human induced pluripotent stem cell-derived cell transplantation therapies to promote endogenous regeneration.
Collapse
Affiliation(s)
- Stephen Vidman
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Yee Hang Ethan Ma
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Nolan Fullenkamp
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Giles W. Plant
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| |
Collapse
|
2
|
Krawiec A, Pietrasik J, Pietrasik Z, Mikuła-Pietrasik J, Książek K. Unveiling the role of extracellular matrix elements and regulators in shaping ovarian cancer growth and metastasis. Cell Signal 2025; 132:111843. [PMID: 40318796 DOI: 10.1016/j.cellsig.2025.111843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/18/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Epithelial ovarian cancer (EOC) progression is determined by numerous intracellular interactions and the interplay between malignant cells, normal cells, and the tumor acellular microenvironment, formed largely by the extracellular matrix (ECM). The structure and biochemical functioning of various ECM components, along with the activity of agents that regulate ECM remodeling, impact the disease's expansion (adhesion, proliferation, invasion), spread, and response to therapy. It is important to note that the involvement of ECM components and their regulators in the progression of EOC is bidirectional and distinctly depends on a particular tissue context. In certain situations, certain components of the ECM enhance the activity of cancer cells, but in other scenarios, they suppress it. In this review, we summarize the newest knowledge regarding diverse aspects of ECM engagement in EOC pathophysiology and chemotherapy. Moreover, we delineate conditions that exacerbate the pro-cancerous properties of ECM, including diabetes-associated glycation, aging, and cellular senescence. We also explore methods to therapeutically alter the properties of the ECM, which could be beneficial in ovarian cancer prevention and treatment.
Collapse
Affiliation(s)
- Adrianna Krawiec
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str, 60-781 Poznań, Poland.
| | - Joanna Pietrasik
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str, 60-781 Poznań, Poland
| | - Zofia Pietrasik
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str, 60-781 Poznań, Poland
| | - Justyna Mikuła-Pietrasik
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str, 60-781 Poznań, Poland.
| | - Krzysztof Książek
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str, 60-781 Poznań, Poland.
| |
Collapse
|
3
|
Nussinov R, Yavuz BR, Jang H. Tumors and their microenvironments: Learning from pediatric brain pathologies. Biochim Biophys Acta Rev Cancer 2025; 1880:189328. [PMID: 40254040 PMCID: PMC12124968 DOI: 10.1016/j.bbcan.2025.189328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
Early clues to tumors and their microenvironments come from embryonic development. Here we review the literature and consider whether the embryonic brain and its pathologies can serve as a better model. Among embryonic organs, the brain is the most heterogenous and complex, with multiple lineages leading to wide spectrum of cell states and types. Its dysregulation promotes neurodevelopmental brain pathologies and pediatric tumors. Embryonic brain pathologies point to the crucial importance of spatial heterogeneity over time, akin to the tumor microenvironment. Tumors dedifferentiate through genetic mutations and epigenetic modulations; embryonic brains differentiate through epigenetic modulations. Our innovative review proposes learning developmental brain pathologies to target tumor evolution-and vice versa. We describe ways through which tumor pharmacology can learn from embryonic brains and their pathologies, and how learning tumor, and its microenvironment, can benefit targeting neurodevelopmental pathologies. Examples include pediatric low-grade versus high-grade brain tumors as in rhabdomyosarcomas and gliomas.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA.
| | - Bengi Ruken Yavuz
- Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| |
Collapse
|
4
|
Alberts A, Kjaer SK, Søegaard SH, Winther JF, Schmiegelow K, Sopina L, Erdmann F, Hargreave M. Childhood vaccinations and the risk of leukemia: A nationwide cohort study. Int J Cancer 2025; 157:55-63. [PMID: 39821269 DOI: 10.1002/ijc.35338] [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: 10/11/2024] [Revised: 12/19/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025]
Abstract
A protective effect of childhood vaccinations on leukemia risk, particularly acute lymphoblastic leukemia (ALL), has been hypothesized, though findings are inconsistent. We used a nationwide cohort of Danish children born 1997-2018 (n = 1,360,230), to examine associations between childhood vaccinations and leukemia (<20 years) using registry data (follow-up: December 31, 2018). Cox proportional hazard models with age as the underlying time estimated hazard ratios (HRs) for leukemia (any, ALL, acute myeloid [AML], and other), comparing vaccinated with unvaccinated children. We also accessed the effect of each additional vaccine dose. During 14,536,819 person-years, 771 children were diagnosed with leukemia (74% ALL, 16% AML, and 10% other). Any vaccination was associated with an increased HR for ALL (HR: 2.76; 95% CI: 0.66-11.58), compared to unvaccinated children, with a change in HR of 1.01 (95% CI: 0.96-1.05) per dose. The corresponding HRs for any leukemia, AML, and other leukemia were 1.04 (95% CI: 0.50-2.17), 0.67 (95% CI: 0.18-2.59) and 0.29 (95% CI: 0.09-0.99), with a change in HR of 0.97 (95% CI: 0.94-1.02), 0.92 (95% CI: 0.84-1.00, p = .062) and 0.88 (95% CI: 0.78-1.00, p = .044) per dose. No significant associations were found for vaccination types, except for the pneumococcal vaccine which was associated with a decreased risk of other leukemia (HR: 0.32; 95% CI: 0.14-0.74). In six-months lag analyses, no significant associations were observed, and decreased risks were attenuated. The results provide no strong evidence that childhood vaccinations reduce leukemia risk. The limited number of unvaccinated cases and wide confidence intervals suggest cautious interpretation of some findings.
Collapse
Affiliation(s)
- Aya Alberts
- Virus, Lifestyle and Genes, Danish Cancer Institute, Copenhagen, Denmark
| | - Susanne K Kjaer
- Virus, Lifestyle and Genes, Danish Cancer Institute, Copenhagen, Denmark
- Department of Gynecology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Signe H Søegaard
- Department of Hematology, Danish Cancer Institute, Copenhagen, Denmark
| | - Jeanette F Winther
- Childhood Cancer Research Group, Danish Cancer Institute, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University and University Hospital, Aarhus, Denmark
| | - Kjeld Schmiegelow
- Pediatric and Adolescent Medicine, Juliane Marie Center, Rigshospitalet, Copenhagen, Denmark
- Institute of Clinical Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Liza Sopina
- Danish Center for Health Economics, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Friederike Erdmann
- Research Group Aetiology and Inequalities in Childhood Cancer, Division of Childhood Cancer Epidemiology, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Department of Prevention and Evaluation, Leibniz Institute for Prevention Research and Epidemiology-BIPS, Bremen, Germany
| | - Marie Hargreave
- Virus, Lifestyle and Genes, Danish Cancer Institute, Copenhagen, Denmark
| |
Collapse
|
5
|
Wan D, Zhang Q, Yang Z, Zhang X, Xie P, Cheng S, Xu L, Liu B, Zhang K, Zhang W. Engineered oncolytic virus OH2-FLT3L enhances antitumor immunity via dendritic cell activation. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200975. [PMID: 40236993 PMCID: PMC11999459 DOI: 10.1016/j.omton.2025.200975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/17/2025] [Accepted: 03/18/2025] [Indexed: 04/17/2025]
Abstract
The combination of oncolytic viruses (OVs) with other immunotherapies, such as immunostimulatory therapies, is a current research hotspot; however, optimizing their therapeutic potential remains to be fully explored. Here, we designed a novel oncolytic herpes simplex virus 2 expressing Fms-like tyrosine kinase 3 ligand (OH2-FLT3L), which induces an antitumor cytotoxic T cell immune response by activating dendritic cells (DCs). We found that OH2-FLT3L specifically infects tumor cells, induces immunogenic cell death (ICD), and releases a large number of tumor-specific antigens, which bound to danger signals and facilitated antigenic cross-presentation by DCs, significantly enhancing T cell activation and function. Experimental results showed that OH2-FLT3L significantly increased the proportion of activated DCs, enhanced the antitumor immune response, and effectively converted "cold" tumors into "hot" tumors. In addition, when combined with anti-PD-1 antibody, OH2-FLT3L further enhanced therapeutic efficacy. In conclusion, OH2-FLT3L, as a novel oncolytic virus, demonstrates the potential to enhance antitumor immune responses through DC activation.
Collapse
Affiliation(s)
- Duo Wan
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qi Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhenrong Yang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Xiaoli Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Peipei Xie
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shujun Cheng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Libin Xu
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Binlei Liu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, College of Bioengineering, Hubei University of Technology, Wuhan 430068, China
| | - Kaitai Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wen Zhang
- Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
6
|
Xiao H, Xu Q, Gao Y, Wu W, Wang B, Li H, Fei M. Identification of CENPW as a prognostic biomarker and potential therapeutic target for clear cell renal cell carcinoma. Discov Oncol 2025; 16:1138. [PMID: 40531390 PMCID: PMC12176707 DOI: 10.1007/s12672-025-02859-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 05/29/2025] [Indexed: 06/22/2025] Open
Abstract
Centromere protein W (CENP-W) is essential for chromosome segregation and mitotic assembly and has been recognized as a prognostic marker in several cancers. However, its significance in clear-cell renal cell carcinoma (ccRCC) remains underexplored. To investigate this, we analyzed transcriptomic data from the National Center for Biotechnology Information (NCBI) and The Cancer Genome Atlas (TCGA) to evaluate CENP-W expression and its associations with clinical outcomes, prognosis, and immune-related markers. Kaplan-Meier survival analysis indicated that elevated CENP-W levels are significantly associated with poorer overall survival in ccRCC patients. Further meta- and multivariate analyses confirmed CENP-W as an independent negative prognostic factor. Gene Set Enrichment Analysis (GSEA) revealed the involvement of CENP-W in immune-related pathways, notably PI3K-Akt and Wnt signaling. Pearson correlation analysis revealed strong associations between CENP-W expression and immune cell infiltration, cancer-associated fibroblasts (CAFs), CTLA4, and PDCD1. qRT-PCR assays confirmed elevated CENP-W levels in ccRCC samples. Additionally, GSEA and GO enrichment highlighted a relationship between CENP-W and lipid metabolism, with reduced CENP-W expression leading to a significant decrease in lipid droplet accumulation. This study identifies CENP-W as a potential biomarker and prognostic indicator in ccRCC, offering insights into personalized therapeutic strategies integrating tumor immunity to enhance the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Haibing Xiao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, People's Republic of China.
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, People's Republic of China.
| | - Qili Xu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Yu Gao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Weikang Wu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Baojun Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Haolin Li
- Department of Urology, The 1st Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| | - Mintian Fei
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, People's Republic of China.
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, People's Republic of China.
| |
Collapse
|
7
|
Araujo M, Bouassaly J, Farshadi F, Hier M, Mascarella M, Mlynarek A, Alaoui-Jamali M, da Silva SD. Current status of circulating tumor DNA and circulating cell alterations in HPV-associated head and neck cancer. Oral Oncol 2025; 167:107417. [PMID: 40516153 DOI: 10.1016/j.oraloncology.2025.107417] [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: 08/21/2024] [Revised: 05/30/2025] [Accepted: 05/31/2025] [Indexed: 06/16/2025]
Abstract
Head and neck cancer (HNC) is the sixth most prevalent cancer worldwide with a poor prognosis when diagnosed at advanced clinical stages. The main risk factors are tobacco consumption and alcohol abuse. However, the incidence of oropharyngeal cancer (OPC) is increasing due to human papillomavirus (HPV) infection. Current diagnostic techniques for both HPV-positive and HPV-negative HNC often involve invasive, costly, and time intensive procedures. Alternatively, liquid biopsies have emerged as a minimally invasive technique which may lessen the burden of cancer diagnoses on both patients and healthcare resources. This technique analyzes biological components released by tumors into the bloodstream, such as circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), exosomes, tumor proteins, and methylation changes, allowing for specific cancer detection and surveillance. This article reviewed the status and clinical applications of ctDNA and CTCs in the diagnosis and treatment of HPV-positive HNC. In addition, a systematic review following PRISMA 2020 Checklist (PROSPERO ID: 560498) was conducted to investigate whether liquid biopsies could be leveraged to assess the role of the immune system on treatment outcomes and the overall survival of HNC patients. Two public databases (Medline and Embase) were searched using relevant MeSH (Medical Subject Headings) terms and keywords. After multiple rounds of screening, eight studies published between 2017 and 2024 involving 814 cancer patients from three different countries were retained for data extraction. The data demonstrated that the immune microenvironment of HNC patients could be characterized via liquid biopsy, however, future validation is required. Furthermore, through the detection of HPV ctDNA, liquid biopsy technology has shown promise in diagnostics, as a predictor of patient prognoses and treatment responses, and as a tool to monitor disease progression in HPV-positive HNC.
Collapse
Affiliation(s)
- Megan Araujo
- Department Medicine, Division of Experimental Medicine - Faculty of Medicine, McGill University, Montreal, QC, Canada; Lady Davis Institute for Medical Research and Segal Cancer Center, Sir Mortimer B. Davis-Jewish General Hospital, Departments of Medicine, Oncology, and Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Jenna Bouassaly
- Department Medicine, Division of Experimental Medicine - Faculty of Medicine, McGill University, Montreal, QC, Canada; Lady Davis Institute for Medical Research and Segal Cancer Center, Sir Mortimer B. Davis-Jewish General Hospital, Departments of Medicine, Oncology, and Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Fatemeh Farshadi
- Department Medicine, Division of Experimental Medicine - Faculty of Medicine, McGill University, Montreal, QC, Canada; Lady Davis Institute for Medical Research and Segal Cancer Center, Sir Mortimer B. Davis-Jewish General Hospital, Departments of Medicine, Oncology, and Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Michael Hier
- Department of Otolaryngology Head and Neck Surgery, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Marco Mascarella
- Department of Otolaryngology Head and Neck Surgery, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, QC, Canada; Lady Davis Institute for Medical Research, Centre for Clinical Epidemiology, Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Alex Mlynarek
- Department of Otolaryngology Head and Neck Surgery, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Moulay Alaoui-Jamali
- Department Medicine, Division of Experimental Medicine - Faculty of Medicine, McGill University, Montreal, QC, Canada; Lady Davis Institute for Medical Research and Segal Cancer Center, Sir Mortimer B. Davis-Jewish General Hospital, Departments of Medicine, Oncology, and Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Sabrina Daniela da Silva
- Department Medicine, Division of Experimental Medicine - Faculty of Medicine, McGill University, Montreal, QC, Canada; Lady Davis Institute for Medical Research and Segal Cancer Center, Sir Mortimer B. Davis-Jewish General Hospital, Departments of Medicine, Oncology, and Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, QC, Canada; Department of Otolaryngology Head and Neck Surgery, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, QC, Canada.
| |
Collapse
|
8
|
Li C, Lin Y, Zheng H, Zeng H, Xu L, Wu D, Lao J, Cai P, Liang S, Wang C, Lin T, Zhong W. Glutamate transporter SLC1A6 promotes resistance to immunotherapy in cancer. Cancer Immunol Immunother 2025; 74:240. [PMID: 40481876 PMCID: PMC12145344 DOI: 10.1007/s00262-025-04074-4] [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: 03/05/2025] [Accepted: 04/30/2025] [Indexed: 06/11/2025]
Abstract
BACKGROUND Resistance to immune checkpoint inhibitors remains a significant challenge in the treatment of cancer. Emerging evidence suggests that metabolic reprogramming plays a crucial role in tumor metabolism and progression. Our study strived to investigate the role and underlying mechanisms of the glutamate transporter SLC1A6 in resistance to immunotherapy of cancer. METHODS Single-cell RNA sequencing was performed on bladder cancer patients receiving neoadjuvant immunotherapy to identify the expression of SLC1A6 in treatment-resistant cases. The clinical prognostic value of SLC1A6 in cancer was validated using publicly available lung cancer single-cell datasets, as well as transcriptomic data from both bladder and lung cancer cohorts. Flow cytometry was employed to assess the impact of SLC1A6 knockdown on the effector function of CD8⁺ T cell. In vivo tumor models were used to evaluate the role of SLC1A6 in immunotherapy resistance, with immunofluorescence staining performed to examine GZMB⁺ CD8⁺ T cell infiltration. RESULTS SLC1A6 was highly expressed in bladder cancer patients resistant to neoadjuvant immunotherapy, and its expression was associated with disease progression, poor prognosis, and low immune infiltration. Knockdown of SLC1A6 in tumor cells enhanced CD8⁺ T cell effector function. SLC1A6 knockdown also improved the efficacy of immunotherapy and increased the infiltration of GZMB⁺ CD8⁺ T cells within the tumor microenvironment. CONCLUSIONS SLC1A6 plays a critical role in resistance to immunotherapy in cancer. Targeting SLC1A6 may provide a promising therapeutic strategy for improving responses to neoadjuvant immunotherapy and advancing combination treatment approaches.
Collapse
Affiliation(s)
- Chenchen Li
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Yi Lin
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Haoran Zheng
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Hengda Zeng
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, People's Republic of China
| | - Longhao Xu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Daqin Wu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Jianwen Lao
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Peicong Cai
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Shuai Liang
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Chunhui Wang
- Department of Urology, Yan'an Hospital Affiliated With Kunming Medical University, Kunming, People's Republic of China
| | - Tianxin Lin
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China.
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, People's Republic of China.
| | - Wenlong Zhong
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
9
|
Mariean CR, Tiuca OM, Mariean A, Cotoi OS. Variation in CBC-Derived Inflammatory Biomarkers Across Histologic Subtypes of Lung Cancer: Can Histology Guide Clinical Management? Diagnostics (Basel) 2025; 15:1437. [PMID: 40507010 PMCID: PMC12155243 DOI: 10.3390/diagnostics15111437] [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: 04/06/2025] [Revised: 05/31/2025] [Accepted: 06/01/2025] [Indexed: 06/16/2025] Open
Abstract
Background/Objectives: The early detection of high levels of CBC-derived inflammatory biomarkers and cellular lines, as well as their variations across different histological subtypes of lung cancer, may aid in the early identification of high-risk lung cancer patients and further guide their clinical approach. Methods: A retrospective descriptive study was conducted and included 202 patients diagnosed with lung carcinoma at the Clinical County Hospital Mureș. The main analyzed parameters were the histological subtype and the stage of the tumor at diagnosis, white blood cell counts, and platelet counts, as well as nine CBC-derived inflammatory indexes like neutrophil-to-lymphocyte ratio (NLR), derived neutrophil-to-lymphocyte ratio (d-NLR), monocyte-to-lymphocyte ratio (MLR), platelet-to-lymphocyte ratio (PLR), eosinophil-to-neutrophil ratio (ENR), eosinophil-to-monocyte ratio (EMR), systemic inflammatory index (SII), systemic inflammatory response index (SIRI), and aggregate index of systemic inflammation (AISI). The statistical analysis was performed using the MedCalc software, version 23.0.2. Logarithmic ANOVA was used to compare groups. Normality was tested using the Shapiro-Wilk test. The Chi-square test compared categorical variables, while the independent Mann-Whitney test was used for continuous variables. Results: The inflammatory response increased as disease severity progressed, with NSCLC-NOS being the histological subtype with the most numerous patients outside the normal ranges. Eosinophil count differed significantly across the histologic subtypes of NSCLC, with adenocarcinoma and adenosquamous patients exhibiting the highest values. In adenocarcinoma patients, we observed that NLR and MLR levels increased progressively as the tumor stage advanced. Based on severity, differences were observed across the histological subtypes of lung cancer in stage III patients for ENR, EMR, AISI, eosinophil count, and platelet count, as well as in stage IV patients for AISI, SIRI, and SII. Disease severity impacts the associated inflammatory response in all histologic subtypes of lung cancer to varying degrees. Conclusions: Histological subtype might have a decisive role in shaping the systemic inflammatory profile of lung cancer patients. CBC-derived indices serve as accessible, cost-effective biomarkers for early risk assessment, aiding in the prognosis evaluation and monitoring of therapeutic response. Future studies are needed to further evaluate the histology-specific inflammatory profiles as adjunctive tools in precision oncology.
Collapse
Affiliation(s)
- Claudia Raluca Mariean
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mureș, 540142 Targu Mureș, Romania
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mureș, 540142 Targu Mureș, Romania
- Department of Radiology, Targu Mureș County Emergency Hospital, 540136 Targu Mureș, Romania
| | - Oana Mirela Tiuca
- Dermatology Department, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mureș, 540142 Targu Mureș, Romania
- Dermatology Clinic, Mures Clinical County Hospital, 540342 Targu Mureș, Romania
| | - Alexandru Mariean
- Pulmonology Clinic, Mures Clinical County Hospital, 540103 Targu Mureș, Romania
| | - Ovidiu Simion Cotoi
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mureș, 540142 Targu Mureș, Romania
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mureș, Romania
| |
Collapse
|
10
|
Saravanan L, Mahale A, Gota V, Khandelia P, Kulkarni OP. Necrostatin-1 attenuates oral squamous cell carcinoma by modulating tumour immune response in mice. Fundam Clin Pharmacol 2025; 39:e70008. [PMID: 40222051 DOI: 10.1111/fcp.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Necroptosis has been shown to play an important role in various pathologies, including pancreatic cancer (PDAC). However, its role in the progression of oral cancer (OSCC) remains unclear. OBJECTIVES To determine the expression of key necroptosis pathway markers in an OSCC mouse model and evaluate the therapeutic effect of a necroptosis inhibitor on the progression of OSCC. METHODS AND RESULTS 4-NQO-induced OSCC in mice resembles very closely to human OSCC. The expression of RIPK-1, RIPK-3, MLKL and their respective phosphorylation was increased in OSCC tissues of cancer-bearing mice. In the analysis of the necroptosis pathway in human OSCC with the TCGA database, we found similar overexpression of RIPK-1 in human cancer, which correlated with the severity of cancer in terms of different cancer grades and stages. Pharmacological blockade of necroptosis with necrostatin-1 (NEC-1) reduced the progression and development of OSCC, characterized by reduced number and severity of tumour lesions, improved histology with reduced hyperplasia, dysplasia and invasive carcinoma. Immune profiling of blood, spleen and tumour tissues demonstrated suppressed expression of MDSCs (CD11b+Gr-1+) and M2-macrophages (CD11b+F4/80+CD206+), while M1-macrophages (CD11b+F4/80+MHCII+) were elevated in the treatment group. The ratio of M2/M1 was reduced in the treated group, suggesting the promotion of anti-tumour immune response. Expression of Arg-1, YM1/2, IL-10 and TGF-β was reduced in tumour tissues in the treated group. CONCLUSION In summary, blocking the necroptosis pathway alters the tumour microenvironment (TME) and inhibits the progression of OSCC. Targeting necroptosis could be an effective therapy for treating OSCC in a clinical setup.
Collapse
Affiliation(s)
- Lavanya Saravanan
- Metabolic and Neuroscience Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Ashutosh Mahale
- Metabolic and Neuroscience Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Vikram Gota
- Advance Centre for Treatment Research & Education in Cancer, Tata Memorial Centre (ACTREC), Navi Mumbai, Maharashtra, India
| | - Piyush Khandelia
- Department of Biological Sciences, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Onkar Prakash Kulkarni
- Metabolic and Neuroscience Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| |
Collapse
|
11
|
Basnet A, Thomas DD, Landreth KM, Damron FH, Liu TW. Immune Response to Bioluminescence Imaging Reporters in Murine Tumor Models. Mol Imaging Biol 2025; 27:341-352. [PMID: 40234300 PMCID: PMC12162783 DOI: 10.1007/s11307-025-02010-7] [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: 03/04/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 04/17/2025]
Abstract
PURPOSE Imaging reporters have been widely employed in cancer research to monitor real-time tumor burden and metastatic spread. These tools offer a valuable approach for non-invasive imaging of tumor dynamics over time. With the established understanding that tumor immunology plays a critical role in cancer progression, it is essential to ensure that the chosen imaging reporters used to study tumor-immune interactions do not inadvertently elicit an immune response. This study aimed to investigate the immune response to bioluminescence reporters used for in vivo tracking of tumor cells in immunocompetent murine models. PROCEDURES The in vitro and in vivo growth effects of two stably expressed bioluminescence reporter genes, a red-shifted firefly luciferase and a click beetle green luciferase, were evaluated in four different cancer cell lines. Differences in parental and reporter-expressing cancer cell immune cell composition, activation, and secreted cytokine levels were evaluated using flow cytometry, cytokine arrays and ELISAs. RESULTS The data revealed no significant differences in in vitro cell proliferation between parental and reporter cancer cell lines. In vivo subcutaneous tumor growth was not observed in tumor cells stably expressing the red-shifted firefly luciferase. Cells labeled with click beetle green luciferase demonstrated no significant differences in in vivo subcutaneous tumor growth compared to parental cells. Tumor cells expressing red-shifted firefly luciferase induced an increase in activated and cytotoxic T cells compared to parental and click beetle green luciferase, suggesting enhanced immunogenicity. Furthermore, the tumor-immune composition and cytokine production were similar between parental and click beetle green luciferase-labeled tumor cells. CONCLUSIONS These findings demonstrate that the stable expression of click beetle green luciferase in cancer cells, in contrast to red-shifted firefly luciferase, has minimal immunogenicity and does not alter tumor development in immunocompetent mice. We report detailed characterization studies of bioluminescence reporter cells, providing essential considerations for their use in investigating tumor-immune interactions in syngeneic murine tumor models.
Collapse
Affiliation(s)
- Angisha Basnet
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA
| | - Dylan D Thomas
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA
| | - Kaitlyn M Landreth
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA
| | - F Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, 26506, USA
| | - Tracy W Liu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA.
- WVU Cancer Institute, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
12
|
Mannan A, Mohan M, Singh TG. Revenge unraveling the fortress: Exploring anticancer drug resistance mechanisms in BC for enhanced therapeutic strategies. Crit Rev Oncol Hematol 2025; 210:104707. [PMID: 40122355 DOI: 10.1016/j.critrevonc.2025.104707] [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: 01/22/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025] Open
Abstract
Breast cancer (BC) is the most prevalent form of cancer in women worldwide and the main cause of cancer-related fatalities in females. BC can be classified into various types based on where cancer has begun to grow or spread, specific characteristics that influence how cancer behaves, and treatment choices. BC is multifaceted, and due to its diverse nature, the mechanisms involved are complex and have not yet been understood. Overexpression and expression of various factors involved in the functioning of mechanisms lead to abnormal changes, providing an environment supporting cancer cell growth. Understanding BC risk factors and early diagnosis through screening techniques like mammography and diagnostic techniques such as imaging and biopsies has advanced significantly. A wide range of treatment options, including surgery, radiation, chemotherapy, targeted treatments, and hormonal therapies, are now available. Daily advancements are being made in the clinical treatment of BC. Still, BC drug resistance cases remain highly prevalent and are currently one of the biggest problems faced by medical science. To increase response rates and possibly lengthen survival, there is a critical requirement for novel medicines with minimal sensitivity to overcome drug resistance. This review classifies different mechanisms that are involved in the development of BC and workable pharmacological targets and explains how they relate to the development of BC drug resistance. By concentrating on the mechanisms covered in this review, we can have a deep understanding of different mechanisms and learn innovative ways to develop novel therapeutics for the disease to combat medication resistance.
Collapse
Affiliation(s)
- Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Maneesh Mohan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
13
|
Li Q, Sheng M, Chen Y, Yi Q, Yang Z, Chen T. Comprehensive immunogenomic landscape analysis unveils CD33 + myeloid cell-driven immunomodulatory signatures in melanoma development. Pathol Res Pract 2025; 270:155981. [PMID: 40300524 DOI: 10.1016/j.prp.2025.155981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/07/2025] [Accepted: 04/16/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND Understanding the causal relationships between immune cell populations and cancer development remains a critical challenge in tumor immunology. METHODS We employed Mendelian Randomization analysis leveraging genome-wide association studies of 612 immune cell traits and 91 cancer types to systematically evaluate causal associations. Single-cell RNA sequencing and computational deconvolution analyses were performed to characterize myeloid cell subpopulations in melanoma samples. FINDINGS Our analysis revealed significant relationships between specific immune cell subsets and cancer risk, particularly highlighting the role of CD33 + myeloid cells in melanoma pathogenesis. Single-cell RNA sequencing identified distinct CD33high myeloid subpopulations characterized by elevated expression of complement cascade components and chemokine signaling pathways. Through computational deconvolution of The Cancer Genome Atlas melanoma cohort, we demonstrated that elevated CD33high monocyte abundance correlates with increased immune dysfunction scores, reduced CD8 + T cell infiltration, and poor survival outcomes. INTERPRETATION Here we delineate the multifaceted mechanisms through which CD33 + myeloid cell populations orchestrate perturbations in the tumor-immune microenvironmental landscape, manifesting in compromised immunosurveillance and enhanced tumor progression. Our findings illuminate novel therapeutic opportunities through targeted modulation of myeloid cell function, while providing a systematic framework for understanding the complex interplay between immune cell populations and oncogenic processes.
Collapse
Affiliation(s)
- Qinke Li
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400010, China; Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Min Sheng
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yiqian Chen
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400010, China; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing 400010, China
| | - Qiang Yi
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400010, China; Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Zhu Yang
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Tong Chen
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400010, China; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing 400010, China.
| |
Collapse
|
14
|
Corica DA, Bell SD, Zhao L, Lawler NJ, Poirier MA, Miller PJ, Wakefield MR, Fang Y. The Era of Precision Medicine: Advancing Treatment Paradigms for Small Cell Lung Cancer. Cancers (Basel) 2025; 17:1847. [PMID: 40507328 PMCID: PMC12153792 DOI: 10.3390/cancers17111847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/29/2025] [Accepted: 05/29/2025] [Indexed: 06/16/2025] Open
Abstract
Small cell lung cancer (SCLC) remains a challenge prognostically. A clinically silent early stage and predilection for early metastasis leads to over half of patients presenting with metastatic disease at the time of diagnosis. Akin to many other cancers, once SCLC metastasizes, current therapies begin to lose their effectiveness. The future of SCLC rests in innovative treatments aimed at improving patient outcomes. Chemotherapy and radiation remain the backbone treatment for SCLC. Most patients diagnosed with SCLC begin treatment with combination chemotherapy consisting of a platinum analog and topoisomerase inhibitor with or without concurrent radiation. Disease progression or recurrence warrants new treatment approaches. New chemotherapy combinations and advances in radiation precision offer patients novel approaches using the same backbone of treatment used in many other cancers. The introduction of newer therapeutic approaches, such as immune checkpoint inhibitors, small molecule targeted therapies, bispecific antibodies, and antibody-drug conjugates offer a bright future for patients with SCLC who fail first-line therapy. This review will focus on advancing treatment paradigms for SCLC in the era of precision medicine. Such a study might be helpful for pulmonologists and oncologists to manage precisely patients with SCLC.
Collapse
Affiliation(s)
- Derek A. Corica
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA; (D.A.C.); (S.D.B.); (N.J.L.); (M.A.P.); (P.J.M.)
| | - Scott D. Bell
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA; (D.A.C.); (S.D.B.); (N.J.L.); (M.A.P.); (P.J.M.)
| | - Lei Zhao
- The Department of Respiratory Medicine, the 2nd People’s Hospital of Hefei and Hefei Hospital Affiliated to Anhui Medical University, Hefei 230002, China;
| | - Nicholas J. Lawler
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA; (D.A.C.); (S.D.B.); (N.J.L.); (M.A.P.); (P.J.M.)
| | - McKade A. Poirier
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA; (D.A.C.); (S.D.B.); (N.J.L.); (M.A.P.); (P.J.M.)
| | - Peyton J. Miller
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA; (D.A.C.); (S.D.B.); (N.J.L.); (M.A.P.); (P.J.M.)
| | - Mark R. Wakefield
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA;
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA; (D.A.C.); (S.D.B.); (N.J.L.); (M.A.P.); (P.J.M.)
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA;
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| |
Collapse
|
15
|
Biswas K, Kuo LS, Shoemaker RH, Mohammed A. Immunoprevention of non-viral cancers: challenges and strategies for early intervention. Cancer Cell Int 2025; 25:196. [PMID: 40437549 PMCID: PMC12121240 DOI: 10.1186/s12935-025-03817-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 05/08/2025] [Indexed: 06/01/2025] Open
Abstract
While the effects of cancer vaccines have been extensively studied in the therapeutic setting, research has been more limited in the areas of cancer prevention and interception. Although cancer prevention by vaccines has been possible for viral-mediated cancers, such as cervical cancers and hepatocellular carcinoma, preventing non-viral cancers by immunopreventive vaccines is challenging. Many tumors at late stages are less responsive to treatments, including immunotherapies and vaccines, in part due to an immunosuppressive microenvironment. Shifting the strategy to intervention at early stages of cancer development and progression and focusing on high-risk cohorts with defined molecular targets offers a pathway for improved vaccine efficacy. Current research on the role of immune mechanisms during tumor initiation and progression is rapidly evolving and recent emerging preclinical immunoprevention studies have shown that vaccines can induce host immune response and effectively control tumor onset and progression. In this review, we address important considerations and challenges regarding the development of cancer immunoprevention for non-viral cancers. We also discuss significant, innovative, and impactful preclinical and clinical immunoprevention studies in various cancers. This includes neoantigen discovery, the use and optimization of immunomodulating agents either alone or in combination with vaccines, and strategies for optimizing vaccines. We conclude by discussing prospects for immunoprevention research and potential opportunities to advance the field in the future.
Collapse
Affiliation(s)
- Kajal Biswas
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD, USA
| | - Lillian S Kuo
- Cancer Immunology, Hematology and Etiology Branch, Division of Cancer Biology, National Cancer Institute, Rockville, MD, USA
| | - Robert H Shoemaker
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD, USA.
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, 9609 Medical Center Drive, Room no: 4E454, Rockville, MD, 20850, USA.
| | - Altaf Mohammed
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD, USA.
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, 9609 Medical Center Drive, Room no: 5E602, Rockville, MD, 20850, USA.
| |
Collapse
|
16
|
Wu Y, Liu J, Yin T, Li X, Liu X, Peng X, Zhan X. SELP can affect the immune microenvironment of gastric cancer and is associated with poor prognosis. Discov Oncol 2025; 16:846. [PMID: 40397261 PMCID: PMC12095770 DOI: 10.1007/s12672-025-02629-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 05/08/2025] [Indexed: 05/22/2025] Open
Abstract
The tumor microenvironment (TME) plays a crucial role in the occurrence and progression of gastric cancer. Yet, we still don't understand how immune and stromal components of TMEs are modulated. In this study, we applied the ESTIMATE algorithm to calculate the number of immune and stromal components in 410 STAD cases in the Cancer Genome Atlas (TCGA) database. COX regression analysis and protein-protein interaction (PPI) network construction were used to analyze differentially expressed genes (DEGs). Then, P-selectin (SELP) was identified as a predictor by cross-analysis of univariate COX and PPI. After verifying the clinical significance of SELP for study, we performed an immune infiltration analysis and identified 54 immunomodulators associated with SELP through public data. Immunomodulation associated with gastric cancer prognosis was then confirmed by LASSO regression, and the previous results were further validated with single-cell data. Finally, we verified that SELP can promote EMT on gastric cancer cells. In conclusion, we validated that SELP may affect the biological phenotype of gastric cancer with the immune microenvironment alteration of gastric cancer.
Collapse
Affiliation(s)
- Yue Wu
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Jingyu Liu
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Tong Yin
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Xiaoxiao Li
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Xian Liu
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Xiaobo Peng
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China.
| | - Xianbao Zhan
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
17
|
Su G, Gao F, Yang M, Wang L, Liang L, Li S, Li G, Han N, Li G, Qian G, Zhang S, Luo H, Zhang D, Liang H, Ren Z. Effectiveness of azvudine versus nirmatrelvir/ritonavir for hospitalized patients with SARS-CoV-2 infection and pre-existing liver diseases. Virol J 2025; 22:147. [PMID: 40389963 PMCID: PMC12087163 DOI: 10.1186/s12985-025-02771-1] [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: 01/17/2025] [Accepted: 05/05/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Azvudine and nirmatrelvir/ritonavir are recommended as priority treatments for SARS-CoV-2 infection in China, but their effectiveness and safety in patients with pre-existing chronic liver diseases remains unknown. METHODS We conducted a multicenter retrospective cohort study of hospitalized SARS-CoV-2 infected patients with chronic liver diseases in ten hospitals of Henan Province. Azvudine recipients were 2:1 propensity score matched with nirmatrelvir/ritonavir recipients. Efficacy and safety were evaluated by Kaplan-Meier analysis, multivariable Cox regression model, subgroup analysis, as well as sensitivity analyses. RESULTS Among 37606 hospitalized patients infected with SARS-CoV-2, 1355 azvudine recipients and 373 nirmatrelvir/ritonavir recipients met the inclusion criteria. Patients with azvudine treatment showed comparable effectiveness to nirmatrelvir/ritonavir with regard to both all-cause death (P = 0.34) and composite disease progression (P = 0.32), even after adjusting for other covariates (all-cause death: HR: 0.80, 95%CI: 0.574-1.128; composite disease progression: HR: 1.31, 95%CI: 0.999-1.723). Notably, compared with nirmatrelvir/ritonavir, azvudine showed better effectiveness for patients with a comorbidity of primary malignant tumor in reducing all-cause death. Four sensitivity analyses further confirmed the robustness. CONCLUSIONS The effectiveness of azvudine may potentially comparable to nirmatrelvir/ritonavir in SARS-CoV-2 infected patients with pre-existing liver diseases with respect to all-cause death and composite disease progression, without serious safety concerns. Due to the existence of potential biases, further studies still need to evaluate the efficacy of these two drugs. TRIAL REGISTRATION The trial was retrospectively registered at ClinicalTrials.gov (CT.gov identifier: NCT06349655).
Collapse
Affiliation(s)
- Guanyue Su
- Department of Infectious Diseases, State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, the First Affiliated Hospital of Zhengzhou University, #1 Jianshe East Road, Zhengzhou, 450052, China
| | - Feng Gao
- Department of Infectious Diseases, State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, the First Affiliated Hospital of Zhengzhou University, #1 Jianshe East Road, Zhengzhou, 450052, China
| | - Mengzhao Yang
- Department of Infectious Diseases, State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, the First Affiliated Hospital of Zhengzhou University, #1 Jianshe East Road, Zhengzhou, 450052, China
| | - Ling Wang
- Department of Clinical Laboratory, Henan Provincial Chest Hospital Affiliated of Zhengzhou University, Zhengzhou, 450008, China
| | - Lili Liang
- Department of Medical Information, Henan Provincial Chest Hospital Affiliated of Zhengzhou University, Zhengzhou, 450008, China
| | - Silin Li
- Department of Respiratory and Critical Care Medicine, Fengqiu County People's Hospital, Xinxiang, 453300, China
| | - Guangming Li
- Department of Liver Disease, the Affiliated Infectious Disease Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Na Han
- Department of Infectious Diseases, State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, the First Affiliated Hospital of Zhengzhou University, #1 Jianshe East Road, Zhengzhou, 450052, China
| | - Guotao Li
- Department of Infectious Diseases, Luoyang Central Hospital Affiliated of Zhengzhou University, Luoyang, 471000, China
| | - Guowu Qian
- Department of Gastrointestinal Surgery, Nanyang Central Hospital, Nanyang, 473009, China
| | - Shixi Zhang
- Department of Infectious Diseases, Shangqiu Municipal Hospital, Shangqiu, 476000, Henan Province, China
| | - Hong Luo
- Guangshan County People's Hospital, Guangshan County, Xinyang, 465450, China
| | - Donghua Zhang
- Department of Infectious Diseases, Anyang City Fifth People's Hospital, Anyang, 455000, China
| | - Hongxia Liang
- Department of Infectious Diseases, State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, the First Affiliated Hospital of Zhengzhou University, #1 Jianshe East Road, Zhengzhou, 450052, China.
| | - Zhigang Ren
- Department of Infectious Diseases, State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, the First Affiliated Hospital of Zhengzhou University, #1 Jianshe East Road, Zhengzhou, 450052, China.
| |
Collapse
|
18
|
Perez-Medina M, Benito-Lopez JJ, Aguilar-Cazares D, Lopez-Gonzalez JS. A Comprehensive Review of Long Non-Coding RNAs in the Cancer-Immunity Cycle: Mechanisms and Therapeutic Implications. Int J Mol Sci 2025; 26:4821. [PMID: 40429961 PMCID: PMC12111859 DOI: 10.3390/ijms26104821] [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: 02/19/2025] [Revised: 05/10/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) have emerged as pivotal regulators of the dynamic interplay between cancer progression and immune responses. This review explored their influence on key processes of the cancer-immunity cycle, such as immune cell differentiation, antigen presentation, and tumor immunogenicity. By modulating tumor escape from the immune response, therapeutic resistance, and tumor-stroma interactions, lncRNAs actively shape the tumor microenvironment. Due to their growing knowledge in the area of immune suppression, directly intervening in the induction of regulatory T cells (Tregs), M2 macrophages, and regulating immune checkpoint pathways such as PD-L1, CTLA-4, and others, lncRNAs can be considered promising therapeutic targets. Advances in single-cell technologies and immunotherapy have significantly expanded our understanding of lncRNA-driven regulatory networks, paving the way for novel precision medicine approaches. Ultimately, we discussed how targeting lncRNAs could enhance cancer immunotherapy, offering new avenues for biomarker discovery and therapeutic intervention.
Collapse
Affiliation(s)
- Mario Perez-Medina
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City 14080, Mexico; (M.P.-M.); (J.J.B.-L.); (D.A.-C.)
- Asociación Para Evitar la Ceguera en México, I. A. P., Mexico City 04030, Mexico
| | - Jesus J. Benito-Lopez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City 14080, Mexico; (M.P.-M.); (J.J.B.-L.); (D.A.-C.)
| | - Dolores Aguilar-Cazares
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City 14080, Mexico; (M.P.-M.); (J.J.B.-L.); (D.A.-C.)
| | - Jose S. Lopez-Gonzalez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City 14080, Mexico; (M.P.-M.); (J.J.B.-L.); (D.A.-C.)
| |
Collapse
|
19
|
Feijó M, Carvalho TMA, Fonseca LRS, Vaz CV, Pereira BJ, Cavaco JEB, Maia CJ, Duarte AP, Kiss-Toth E, Correia S, Socorro S. Endocrine-disrupting chemicals as prostate carcinogens. Nat Rev Urol 2025:10.1038/s41585-025-01031-9. [PMID: 40379948 DOI: 10.1038/s41585-025-01031-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2025] [Indexed: 05/19/2025]
Abstract
Endocrine-disrupting chemicals (EDCs) are natural or synthetic compounds that are ubiquitous in the environment and in daily-usage products and interfere with the normal function of the endocrine system leading to adverse health effects in humans. Exposure to these chemicals might elevate the risk of metabolic disorders, developmental and reproductive defects, and endocrine-related cancers. Prostate cancer is the most common hormone-dependent cancer in men, and the fifth leading cause of cancer-related mortality, partly owing to a lack of knowledge about the mechanisms that lead to aggressive castration-resistant forms. In addition to the dependence of early-stage prostate cancer on androgen actions, the prostate is a target of oestrogenic regulation. This hormone dependence, along with the fact that exogenous influences are major risk factors for prostate cancer, make the prostate a likely target of harmful actions from EDCs. Various sources of EDCs and their different modes of action might explain their role in prostate carcinogenesis.
Collapse
Affiliation(s)
- Mariana Feijó
- RISE-Health, Department of Chemistry, Faculty of Sciences, University of Beira Interior, Covilhã, Portugal
| | - Tiago M A Carvalho
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Lara R S Fonseca
- RISE-Health, Department of Chemistry, Faculty of Sciences, University of Beira Interior, Covilhã, Portugal
| | - Cátia V Vaz
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Bruno J Pereira
- Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
- Instituto Português de Oncologia de Coimbra, Coimbra, Portugal
| | - José Eduardo B Cavaco
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Cláudio J Maia
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Ana P Duarte
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Endre Kiss-Toth
- School of Medicine and Population Health, University of Sheffield, Sheffield, United Kingdom
| | - Sara Correia
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.
| | - Sílvia Socorro
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|
20
|
Zhou D, Sun Y, Ding P, Wang X, Li L, Li L, Lv X, Liao T, Chen J, Zhang W, Wang Q, Ji QH, Gao F, Hu W. Gut commensal bacteria influence colorectal cancer development by modulating immune response in AOM/DSS-treated mice. Microbiol Spectr 2025:e0279224. [PMID: 40377337 DOI: 10.1128/spectrum.02792-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/30/2025] [Indexed: 05/18/2025] Open
Abstract
The gut microbiota has been closely associated with the pathogenesis of colorectal cancer (CRC). However, precise identification of particular microorganisms promoting CRC carcinogenesis, and more importantly those blocking tumor development, has been challenging based on human gut microbiota profiling studies. With a well-established azoxymethane/dextran sodium sulfate induction murine CRC model, we found a subset of mice consistently failed to develop CRC. This genetically homogeneous but cancer-refractory population gave us a unique opportunity to reveal that the microbial compositions between mice with and without CRC formation are indeed distinct, indicating key different gut microbiota between those groups are responsible for the differential susceptibility of the animals to CRC development. Our analysis revealed that Ruminococcus flavefaciens (R.f) and Fibrobacter succinogenes (F.s) were significantly enriched in CRC-free mice, while the presence of Eubacterium dolichum (E.d) was dramatically reduced. The correlative evidence was further substantiated as important causal factors, with subsequent bacteria intragastric administration experiments demonstrating independent, protective roles of R.f and F.s and a correspondingly detrimental role of E.d in inflammation-induced CRC initiation. Notably, E.d strongly activates NF-κB and promotes the local accumulation of myeloid-derived suppressor cells and macrophages. Significant disturbance of gut immune homeostasis, therefore, might be a critical trigger leading to subsequent CRC development. These findings indicate a clear direction for precise and rational gut microbiota-mediated CRC prevention.IMPORTANCEThere is a complex ecosystem of different microbes residing within the gut, which is highly relevant to health and diseases. The causal linkage between specific gut microbes and the development of colorectal cancer has been established with a mouse model, pinpointing specific bacteria species either promoting or preventing colorectal cancer development. A key aspect of these gut residual bacteria in colorectal cancer development is through exaggerating or easing gut inflammation. Therefore, by taking probiotics composed of corresponding cancer-preventing bacteria from human microbiota, it can be an effective and economic way to reduce human colorectal cancer risks.
Collapse
Affiliation(s)
- Danlei Zhou
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yujing Sun
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peipei Ding
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaochao Wang
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ling Li
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Luying Li
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinyue Lv
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tian Liao
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Zhengzhou, China
| | - Jianfeng Chen
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Zhang
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qi Wang
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qing-Hai Ji
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Zhengzhou, China
| | - Feng Gao
- Department of Neuroimmunology, Zhengzhou University, Zhengzhou, Henan, China
- Henan Engineering Technology Research Center for Accurate Diagnosis Neuroimmunity, Zhengzhou University, Zhengzhou, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Weiguo Hu
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
- Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Peng G, Li B, Han H, Yuan Y, Mishra F, Huang Y, Liu ZR. Extracellular PKM2 modulates cancer immunity by regulating macrophage polarity. Cancer Immunol Immunother 2025; 74:195. [PMID: 40343475 PMCID: PMC12064527 DOI: 10.1007/s00262-025-04050-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: 11/06/2024] [Accepted: 04/10/2025] [Indexed: 05/11/2025]
Abstract
Tumor controls its immunity by educating its microenvironment, including regulating polarity of tumor associated macrophages. It is well documented that cancer cells release PKM2 to facilitate tumor progression. We report here that the extracellular PKM2 (EcPKM2) modulates tumor immunity by facilitating M2 macrophage polarization in tumors. EcPKM2 interacts with integrin αvβ3 on macrophage to activate integrin-FAK-PI3K signal axis. Activation of FAK-PI3K by EcPKM2 suppresses PTEN expression, which subsequently upregulates arginase1 (Arg1) expression and activity in macrophage to facilitate M2 polarity. Our studies uncover a novel and important mechanism for modulation of tumor immunity. More importantly, an antibody against PKM2 that disrupts the interaction between EcPKM2 and integrin αvβ3 is effective in converting M2 macrophages to M1 macrophages in tumors, suggesting a new therapeutic strategy and target for cancer therapies. Combination of the anti-PKM2 antibody with checkpoint blockades provides enhanced treatment effects.
Collapse
Affiliation(s)
- Guangda Peng
- Department of Biology, Georgia State University, University Plaza, Atlanta, GA, 30303, USA
| | - Bin Li
- Department of Biology, Georgia State University, University Plaza, Atlanta, GA, 30303, USA
| | - Hongwei Han
- Department of Biology, Georgia State University, University Plaza, Atlanta, GA, 30303, USA
| | - Yi Yuan
- Department of Biology, Georgia State University, University Plaza, Atlanta, GA, 30303, USA
| | - Falguni Mishra
- Department of Biology, Georgia State University, University Plaza, Atlanta, GA, 30303, USA
| | - Yang Huang
- Department of Biology, Georgia State University, University Plaza, Atlanta, GA, 30303, USA
| | - Zhi-Ren Liu
- Department of Biology, Georgia State University, University Plaza, Atlanta, GA, 30303, USA.
| |
Collapse
|
22
|
Mangiola S, Brown R, Zhan C, Berthelet J, Guleria S, Liyanage C, Ostrouska S, Wilcox J, Merdas M, Fuge-Larsen P, Bell C, Schröder J, Mielke LA, Mariadason JM, Tsao SCH, Chen Y, Yadav VK, Vodala S, Anderson RL, Merino D, Behren A, Yeo B, Papenfuss AT, Pal B. Circulating immune cells exhibit distinct traits linked to metastatic burden in breast cancer. Breast Cancer Res 2025; 27:73. [PMID: 40340807 PMCID: PMC12063295 DOI: 10.1186/s13058-025-01982-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 02/14/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Circulating immune cells play a crucial role in the anti-tumour immune response, yet the systemic immune system in metastatic breast cancers is not fully characterised. Investigating the cellular and molecular changes in peripheral blood mononuclear cells (PBMCs) from breast cancer patients could elucidate the role of circulating immune cells in metastasis and aid in identifying biomarkers for disease burden and progression. METHODS In this study, we characterised the systemic immune landscape associated with varying levels of metastatic burden by analysing the single-cell transcriptomes of PBMCs from breast cancer patients and healthy controls. Our research focused on identifying changes in immune cell composition, transcriptional programs, and immune-cell communication networks linked to metastatic burden. Additionally, we compared these PBMC features onto a single-cell atlas of primary breast tumours to study corresponding traits in tumour-infiltrating immune cells. RESULTS In metastatic breast cancer, PBMCs exhibit a significant downregulation of the adaptive immune system and a decreased number and activity of unconventional T cells, such as γδ T cells. Additionally, metastatic burden is associated with impaired cell communication pathways involved in immunomodulatory functions. We also identified a gene signature derived from myeloid cells shared between tumour immune infiltrates and circulating immune cells in breast cancer patients. CONCLUSIONS Our study provides a comprehensive single-cell molecular profile of the peripheral immune system in breast cancer, offering a valuable resource for understanding metastatic disease in terms of tumour burden. By identifying immune traits linked to metastasis, we have unveiled potential new biomarkers of metastatic disease.
Collapse
Affiliation(s)
- S Mangiola
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia.
- South Australian immunoGENomics Cancer Institute, Adelaide, SA, 5005, Australia.
| | - R Brown
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - C Zhan
- South Australian immunoGENomics Cancer Institute, Adelaide, SA, 5005, Australia
| | - J Berthelet
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - S Guleria
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - C Liyanage
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - S Ostrouska
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - J Wilcox
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - M Merdas
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - P Fuge-Larsen
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - C Bell
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
- Austin Health, Heidelberg, VIC, 3084, Australia
| | - J Schröder
- Peter Doherty Institute for Infection and Immunity, Parkville, VIC, 3052, Australia
- The University of Melbourne, Parkville, VIC, 3052, Australia
| | - L A Mielke
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - J M Mariadason
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - S Chang-Hao Tsao
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
- Austin Health, Heidelberg, VIC, 3084, Australia
| | - Y Chen
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - V K Yadav
- Rutgers New Jersey Medical School, Newark, NJ, USA
| | - S Vodala
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, USA
| | - R L Anderson
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - D Merino
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - A Behren
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - B Yeo
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
- Austin Health, Heidelberg, VIC, 3084, Australia
| | - A T Papenfuss
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia.
| | - B Pal
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia.
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.
| |
Collapse
|
23
|
Shu X, Sun J, Zhang M, Li X, Yang N, Sun X, Wang X, Guo X, Buas M, Watt G, Li X. Disentangling the Complex Relationships Between Autoimmune Diseases and Cancer Through Polygenic Risk Scores: Evidence from a Large Prospective Study. RESEARCH SQUARE 2025:rs.3.rs-6491161. [PMID: 40386413 PMCID: PMC12083696 DOI: 10.21203/rs.3.rs-6491161/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/02/2025]
Abstract
Although prior studies have reported a link between autoimmunity and carcinogenesis, the roles of genetic factors and potential mediators involved in this process remain elusive. We constructed disease-specific and combined polygenic risk scores (PRSs) for 6 common autoimmune diseases (AIDs) (systemic lupus erythematosus [SLE], rheumatoid arthritis, multiple sclerosis, Crohn's disease, ulcerative colitis [UC], and type 1 diabetes mellitus [T1D]) in the UK Biobank cohort. Compared to those without AID, AID patients at baseline had increased risks of overall cancer, hematological, digestive, and urinary cancer. The combined AID-PRS was significantly associated with increased risks of hematological cancer (HR [95% CI] per SD increase: 1.06 [1.03-1.09]) and non-Hodgkin's lymphoma (HR [95% CI] per SD increase: 1.10 [1.05-1.14]). For individual AID-PRSs, we identified 21 significant associations between 5 PRSs and 11 types of cancer in the overall population, along with 15 additional associations in the sex-stratified analysis. For example, SLE-, UC-, and T1D-PRS showed complex cross-cancer effects on risks of up to 6 cancer types. These associations were generally independent of immunosuppressant drug use. Differential associations of SLE-PRS with prostate cancer risk were found by prostate cancer PRS status ( P interaction <0.05). Several peripheral biomarkers, including red or white blood cell counts, platelet counts, and CRP partly mediated the PRS associations (up to 16.96%). Our study provides important insights into the role of autoimmune diseases in carcinogenesis, which also highlights opportunities for target cancer screening and prevention in potentially vulnerable populations.
Collapse
|
24
|
Aquino A, Franzese O. Reciprocal Modulation of Tumour and Immune Cell Motility: Uncovering Dynamic Interplays and Therapeutic Approaches. Cancers (Basel) 2025; 17:1547. [PMID: 40361472 PMCID: PMC12072109 DOI: 10.3390/cancers17091547] [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: 03/31/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
Dysregulated cell movement is a hallmark of cancer progression and metastasis, the leading cause of cancer-related mortality. The metastatic cascade involves tumour cell migration, invasion, intravasation, dissemination, and colonisation of distant organs. These processes are influenced by reciprocal interactions between cancer cells and the tumour microenvironment (TME), including immune cells, stromal components, and extracellular matrix proteins. The epithelial-mesenchymal transition (EMT) plays a crucial role in providing cancer cells with invasive and stem-like properties, promoting dissemination and resistance to apoptosis. Conversely, the mesenchymal-epithelial transition (MET) facilitates metastatic colonisation and tumour re-initiation. Immune cells within the TME contribute to either anti-tumour response or immune evasion. These cells secrete cytokines, chemokines, and growth factors that shape the immune landscape and influence responses to immunotherapy. Notably, immune checkpoint blockade (ICB) has transformed cancer treatment, yet its efficacy is often dictated by the immune composition of the tumour site. Elucidating the molecular cross-talk between immune and cancer cells, identifying predictive biomarkers for ICB response, and developing strategies to convert cold tumours into immune-active environments is critical to overcoming resistance to immunotherapy and improving patient survival.
Collapse
Affiliation(s)
| | - Ornella Franzese
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
| |
Collapse
|
25
|
Puri N, Sahane P, Phatale V, Khairnar P, Shukla S, Priyadarshinee A, Jain A, Srivastava S. Nano-chameleons: A review on cluster of differentiation-driven immune cell-engineered nanoarchitectonics for non-small cell lung cancer. Int J Biol Macromol 2025; 310:143440. [PMID: 40280523 DOI: 10.1016/j.ijbiomac.2025.143440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/26/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
Cancer, being one of the most outrageous diseases, contributed to 48 % of the mortality in 2022, with lung cancer leading the race with a 12.4 % incidence rate. Conventional treatment modalities like radio-, chemo-, photo-, and immunotherapy employing nanocarriers often face several setbacks, such as non-specific delivery, off-site toxicity, rapid opsonization via the host immune system, and greater tumor recurrence rates. Moreover, the heterogeneous variability in the tumor microenvironment is responsible for existing therapy failure. With the advent of biomimetic nanoparticles as a novel and intriguing platform, researchers have exploited the inherent functionalities of the Cluster of Differentiation proteins (CD) as cell surface biomarkers and imparted the nanocarriers with enhanced homologous tumor targetability, immune evasion capability, and stealth properties, paving the way for improved therapy and diagnosis. This article explores pathogenesis and the multifaceted role of immune cells in non-small cell lung cancer. Moreover, the agenda of this article is to shed light on biomimetic nanoarchitectonics with respect to their fabrication, evaluation, and applications unraveling their synergistic effect with conventional therapies. Further discussion mentions the hurdles in clinical translation with viable solutions. The regulatory bottlenecks underscore the need for a regulatory roadmap with respect to commercialization. We believe that biomimetic nanoarchitectonics will be a beacon of hope in warfare against lung cancer.
Collapse
Affiliation(s)
- Niharika Puri
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Prajakta Sahane
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Vivek Phatale
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Pooja Khairnar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Shalini Shukla
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Abhipsa Priyadarshinee
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Akshita Jain
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India.
| |
Collapse
|
26
|
Tian S, Liu Y, Tan Y, Cui X, Liu R, Liu C, Zhao Y, Xu K, Zhou J. Necroptosis-inducing nanobubbles for effective oxygen delivery and enhanced sonodynamic immunotherapy of breast cancer via UTND. Eur J Pharm Biopharm 2025; 210:114675. [PMID: 39993510 DOI: 10.1016/j.ejpb.2025.114675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
PURPOSE Breast cancer (BC) is a global threat to female health. Sonodynamic therapy (SDT) has been shown to induce apoptosis in tumor cells and trigger immunogenic cell death, leading to the activation of antitumor immunity. However, the immunogenicity of this process may be compromised by oxidative stress and proteolysis. Necroptosis caused by ultrasound-targeted nanobubble destruction (UTND) could boost immunity. Therefore, we tested if necroptosis-inducible nanobubbles (NB) could enhance sonodynamic immunotherapy for BC. We also assessed whether O2-filled NB could address tumor hypoxia and enhance SDT efficacy. METHODS A novel multifunctional nano-system, comprising NB for UTND encapsulating chlorin e6 (Ce6) for SDT and O2 for hypoxia alleviation was established. Ce6-O2NB cytocompatibility and intracellular uptake was studied in vitro, as well as whether Ce6-O2NB could generated reactive oxygen species when exposed to ultrasound irradiation in order to induce apoptosis in tumor cells. In vivo pharmacokinetics, therapeutic efficacy, and immune activation after Ce6-O2NB treatment were studied in 4T1 tumor-bearing mice. RESULTS Ce6-O2NB had a well-designed core-shell structure and desirable biocompatibility and safe therapeutic effects. Ce6-O2NB was able to load both ce6 and oxygen to increase ce6 and oxygen accumulation in tumors. After triggering by ultrasound, Ce6-O2NB generated reactive oxygen species (ROS) and acted as sonosensitizers for SDT, promoting tumor cell death through apoptotic and/or necrotic mechanisms. Furthermore, antitumor immunity was activated by stimulation of spleen lymphocyte proliferation and cytotoxicity, and increasing cytotoxic T lymphocyte numbers. Combination of oxygen with SDT ultimately strengthened its antitumor effects. In addition, Ce6-O2NB alleviated tumor hypoxia, induced increased ROS generation, and improved immune responses and therapeutic efficacy of SDT. Ce6-O2NB also facilitated fluorescence and contrast-enhanced ultrasound imaging. CONCLUSIONS Ce6-O2NB can mitigate tumor hypoxia, enhance SDT, and activate antitumor immunity by inducing simultaneous immunogenic apoptosis and necroptosis, ultimately activating antitumor immunity and inhibiting breast tumor growth in mice.
Collapse
Affiliation(s)
- Shun Tian
- Department of Ultrasound Imaging, The Second People's Hospital of China Three Gorges University, Yichang 443000, China
| | - Yun Liu
- Department of Ultrasound Imaging, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443008, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 334002, China
| | - Yandi Tan
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xinwu Cui
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rong Liu
- Department of Ultrasound Imaging, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443008, China
| | - Chaoqi Liu
- Medical College, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 334002, China
| | - Yun Zhao
- Medical College, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 334002, China
| | - Kui Xu
- Department of Ultrasound Imaging, The Second People's Hospital of China Three Gorges University, Yichang 443000, China
| | - Jun Zhou
- Department of Ultrasound Imaging, The Second People's Hospital of China Three Gorges University, Yichang 443000, China.
| |
Collapse
|
27
|
Wang H, Herman A, Barrow F, Abdel-Ghani A, Draxler M, Fredrickson G, Parthiban P, Seelig DM, Ikramuddin S, Revelo XS. Single-cell RNA sequencing reveals a reprogramming of hepatic immune cells and a protective role for B cells in MASH-driven HCC. Hepatol Commun 2025; 9:e0668. [PMID: 40257356 PMCID: PMC12014033 DOI: 10.1097/hc9.0000000000000668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 12/11/2024] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND HCC, the most common form of liver cancer, is one of the leading causes of cancer-related deaths worldwide. Although the immune system plays a crucial role in liver cancer pathogenesis, the immune landscape within metabolic dysfunction-associated steatohepatitis-driven HCC remains poorly understood. METHODS In this study, we used the high-fat, high-carbohydrate diet fed major urinary protein-urokinase-type plasminogen activator mouse model of metabolic dysfunction-associated steatohepatitis-driven HCC. We performed single-cell RNA sequencing on intrahepatic immune cells to characterize their heterogeneity and gene expression profiles. Additionally, we examined the role of B cells in antitumor immunity by depleting B cells in μMT mice and analyzing the effects on liver cancer progression. RESULTS Our analysis revealed significant shifts in intrahepatic immune cell populations, including B cells, T cells, and macrophages that undergo transcriptional reprogramming, suggesting altered roles in tumor immunity. Notably, an expanded subset of activated B cells in HCC mice showed an antitumor B cell gene expression signature associated with increased survival of patients with liver cancer. Consistently, B cell-deficient mice showed exacerbated liver cancer progression, a substantial reduction in intrahepatic lymphocytes, and impaired CD8+ T cell activation, suggesting that intrahepatic B cells may promote antitumor immunity by enhancing T cell responses. CONCLUSIONS Our findings reveal a complex immune reprogramming within the metabolic dysfunction-associated steatohepatitis-driven HCC microenvironment and underscore a protective role for B cells in liver cancer. These results highlight B cells as potential targets for immunomodulatory therapies in HCC.
Collapse
Affiliation(s)
- Haiguang Wang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Adam Herman
- Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Fanta Barrow
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Amal Abdel-Ghani
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Micah Draxler
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Gavin Fredrickson
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Preethy Parthiban
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Davis M. Seelig
- Veterinary Clinical Sciences Department, Comparative Pathology Shared Resource, University of Minnesota, Saint Paul, Minnesota, USA
| | - Sayeed Ikramuddin
- Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Xavier S. Revelo
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
- Institute for the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
28
|
Aden D, Zaheer S, Sureka N, Trisal M, Chaurasia JK, Zaheer S. Exploring immune checkpoint inhibitors: Focus on PD-1/PD-L1 axis and beyond. Pathol Res Pract 2025; 269:155864. [PMID: 40068282 DOI: 10.1016/j.prp.2025.155864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 01/20/2025] [Accepted: 02/25/2025] [Indexed: 04/19/2025]
Abstract
Immunotherapy emerges as a promising approach, marked by recent substantial progress in elucidating how the host immune response impacts tumor development and its sensitivity to various treatments. Immune checkpoint inhibitors have revolutionized cancer therapy by unleashing the power of the immune system to recognize and eradicate tumor cells. Among these, inhibitors targeting the programmed cell death protein 1 (PD-1) and its ligand (PD-L1) have garnered significant attention due to their remarkable clinical efficacy across various malignancies. This review delves into the mechanisms of action, clinical applications, and emerging therapeutic strategies surrounding PD-1/PD-L1 blockade. We explore the intricate interactions between PD-1/PD-L1 and other immune checkpoints, shedding light on combinatorial approaches to enhance treatment outcomes and overcome resistance mechanisms. Furthermore, we discuss the expanding landscape of immune checkpoint inhibitors beyond PD-1/PD-L1, including novel targets such as CTLA-4, LAG-3, TIM-3, and TIGIT. Through a comprehensive analysis of preclinical and clinical studies, we highlight the promise and challenges of immune checkpoint blockade in cancer immunotherapy, paving the way for future advancements in the field.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India.
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Monal Trisal
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| |
Collapse
|
29
|
Kamiki J, Gorgulho CM, Lérias JR, Maeurer MJ. Mucosal-associated invariant T-cells in pulmonary pathophysiology. Curr Opin Pulm Med 2025; 31:202-210. [PMID: 40104908 PMCID: PMC11957436 DOI: 10.1097/mcp.0000000000001163] [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] [Indexed: 03/20/2025]
Abstract
PURPOSE OF REVIEW Mucosal-associated invariant T-cells (MAIT) have been associated with lung cancer and pulmonary infections. The treatment of patients with cancer or infections includes host-directed therapies (HDTs). MAIT play a role in shaping the 'milieu interne' in cancer and infections and this review addresses the biology of MAIT in pulmonary pathophysiology. RECENT FINDINGS MAIT represent an attractive target for therapy in pulmonary malignancies and infections. T-cells are often difficult to exploit therapeutically due to the diversity of both T-cell receptor (TCR) repertoire and its ligandome. MAIT-cells are restricted by the major histocompatibility complex class I-related gene protein (MR1) that presents nondefined tumor-associated targets, bacterial products, vitamin and drug derivates. Due to their plasticity in gene expression, MAIT are able to conversely switch from IFN-γ to IL-17 production. Both cytokines play a key role in protective immune responses in infections and malignancies. MAIT-derived production of interleukin (IL)-17/TGF-β shapes the tumor micro-environment (TME), including tissue re-modelling leading to pulmonary fibrosis and recruitment of neutrophils. MAIT contribute to the gut-lung axis associated with clinical improved responses of patients with cancer to checkpoint inhibition therapy. MAIT are at the crossroad of HDTs targeting malignant and infected cells. Clinical presentations of overt inflammation, protective immune responses and tissue re-modeling are reviewed along the balance between Th1, Th2, Th9, and Th17 responses associated with immune-suppression or protective immune responses in infections. SUMMARY MAIT shape the TME in pulmonary malignancies and infections. Drugs targeting the TME and HDTs affect MAIT that can be explored to achieve improved clinical results while curbing overt tissue-damaging immune responses.
Collapse
Affiliation(s)
- Jéssica Kamiki
- ImmunoTherapy/ImmunoSurgery Laboratory, Cell Center at the Champalimaud Foundation, Lisbon, Portugal
| | | | | | | |
Collapse
|
30
|
Eljilany I, Garcia JR, Jamal B, Tarhini AA. Monoclonal antibodies as adjuvant therapies for resected melanoma. Expert Opin Biol Ther 2025; 25:1-14. [PMID: 40125987 DOI: 10.1080/14712598.2025.2484305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 03/25/2025]
Abstract
INTRODUCTION Systemic adjuvant therapy is indicated in patients with high-risk, resected melanoma to reduce recurrence risk and potentially improve survival rates. Monoclonal antibodies (mAbs) target immune checkpoints and have made significant advances as systemic adjuvant therapies. AREAS COVERED This review discusses the main clinical trials that tested adjuvant mAbs in resected high-risk melanoma, including anti-cytotoxic T-lymphocyte antigen-4 (CTLA-4) and anti-programmed cell death-1 (PD-1); in addition to newer immunotherapies being tested in the adjuvant setting, including anti-lymphocyte activation gene 3 (LAG-3). We also briefly discuss targeted therapies as an alternative choice. Moreover, we highlight the pros and cons of using mAbs in the adjuvant setting, the reported adverse events (AEs), and the quality of life impact. Finally, we report data related to biomarker studies tested in the context of these clinical trials. EXPERT OPINION Immune checkpoint inhibitors (ICIs) have been shown to significantly improve relapse-free survival (RFS) as adjuvant therapy for high-risk melanoma. The long-term impact on overall survival (OS) was demonstrated in two trials that tested ipilimumab as compared to placebo (EORTC18071) and interferon-α (ECOG-ACRIN E1609). Furthermore, emerging data with neoadjuvant therapy followed by surgery and adjuvant therapy utilizing ICIs have demonstrated improved outcomes in the management of locoregionally advanced disease when compared to upfront surgery followed by adjuvant therapy alone.
Collapse
Affiliation(s)
- Islam Eljilany
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Julia R Garcia
- Department of Medical Oncology, Beneficência Portuguesa de São Paulo, São Paulo, Brazil
| | - Basmala Jamal
- Department of Health Sciences, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Ahmad A Tarhini
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
31
|
Saeidpour Masouleh S, Nasiri K, Ostovar Ravari A, Saligheh Rad M, Kiani K, Sharifi Sultani A, Nejati ST, Nabi Afjadi M. Advances and challenges in CAR-T cell therapy for head and neck squamous cell carcinoma. Biomark Res 2025; 13:69. [PMID: 40312353 PMCID: PMC12044960 DOI: 10.1186/s40364-025-00783-1] [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: 02/08/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) remains among the most aggressive malignancies with limited treatment options, especially in recurrent and metastatic cases. Despite advances in surgery, radiotherapy, chemotherapy, and immune checkpoint inhibitors, survival rates remain suboptimal due to tumor heterogeneity, immune evasion, and treatment resistance. In recent years, Chimeric Antigen Receptor (CAR) T-cell therapy has revolutionized hematologic cancer treatment by genetically modifying T cells to target tumor-specific antigens like CD19, CD70, BCMA, EGFR, and HER2, leading to high remission rates. Its success is attributed to precise antigen recognition, sustained immune response, and long-term immunological memory, though challenges like cytokine release syndrome and antigen loss remain. Notably, its translation to solid tumors, including HNSCC, faces significant challenges, such as tumor microenvironment (TME)-induced immunosuppression, antigen heterogeneity, and limited CAR T-cell infiltration. To address these barriers, several tumor-associated antigens (TAAs), including EGFR, HER2 (ErbB2), B7-H3, CD44v6, CD70, CD98, and MUC1, have been identified as potential CAR T-cell targets in HNSCC. Moreover, innovative approaches, such as dual-targeted CAR T-cells, armored CARs, and CRISPR-engineered modifications, aim to enhance efficacy and overcome resistance. Notably, combination therapies integrating CAR T-cells with immune checkpoint inhibitors (e.g., PD-1/CTLA-4 blockade) and TGF-β-resistant CAR T designs are being explored to improve therapeutic outcomes. This review aimed to elucidate the current landscape of CAR T-cell therapy in HNSCC, by exploring its mechanisms, targeted antigens, challenges, emerging strategies, and future therapeutic potential.
Collapse
Affiliation(s)
| | - Kamyar Nasiri
- Faculty of Dentistry, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Ava Ostovar Ravari
- Faculty of Dentistry, Haybusak University of Medical Sciences, Yerevan, Armenia
| | - Mona Saligheh Rad
- Faculty of Dentistry, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Kiarash Kiani
- Faculty of Dentistry, Islamic Azad University of Medical Sciences, Tehran, Iran
| | | | | | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
32
|
Wu X, Jin Z, Li B, Lu Y, Hou J, Yao L, Yu Z, Sang Q, Yu B, Li J, Li C, Yan C, Zhu Z, Tang K, Liu B, Su L. Deciphering of intra-tumoural heterogeneity and the interplay between metastasis-associated meta-program and myofibroblasts in gastric cancer. Clin Transl Med 2025; 15:e70319. [PMID: 40292733 PMCID: PMC12035649 DOI: 10.1002/ctm2.70319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/28/2025] [Accepted: 04/13/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) exhibits high heterogeneity that relies on the oncogenic properties of cancer cells and multicellular interactions in the tumour microenvironment. However, the heterogeneity of GC and their molecular characteristics are still largely unexplored. METHODS We employed single-cell and spatial transcriptomics to comprehensively map the intra-tumoural heterogeneity within GC. Additionally, in vitro experiments, clinical sample analyses, and patient-derived organoid models (PDOs) were conducted to validate the key interaction patterns between tumor cells and stromal cells. RESULTS Seven robust meta-programs (MP1-MP7) in GC were defined with distinct biological significance and spatial distributions. MP3 and MP4 were intimately associated with distinct CD8 T cells skewed toward a cytotoxic or exhaustion state, while MP7, characterised by the highest degree of malignancy, harboured an immune lockdown microenvironment around it and spatially associated with myofibroblasts (myCAFs). Notably, we clarified the interplay between the MP7 and myCAFs, where MP7 induces the chemotactic migration of fibroblasts and promoting their transformation into myCAFs via GDF15/TGFBR2, and in turn, myCAFs-derived RSPO3 up-regulates EGR1 to promote the transformation to MP7 in GC cells and human PDOs. Ultimately, the accumulation of myCAFs around MP7 led to fewer infiltration of CD8 T cells, resulting an immune-deprived microenvironment and the diminished efficacy of immunotherapy. Additionally, based on the gene expression signatures of MP7 GC cells, we predicted specific drugs and verified more potent inhibitory effects of Taselisib and Lapatinib for MP7 GC cells than conventional drugs at the same concentration. CONCLUSION Taken together, these results deepened the understanding of GC heterogeneity and paved the way for novel therapeutic strategies by targeting MP7 GC cells and their interaction loop with myCAFs in GC treatment. KEY POINTS Seven robust meta-programs (MP1-MP7) were identified in gastric cancer. MP7 was strongly correlated with cancer metastasis and poor survival of gastric cancer patients. MP7 promoted fibroblast transformation into myCAFs via GDF15/TGFBR2, creating an immune lockdown microenvironment. MyCAFs induced MP7 transformation via the RSPO3/EGR1 pathway, promoting gastric cancer cell migration. Taselisib and Lapatinib were potent inhibitors of MP7 GC cells.
Collapse
Affiliation(s)
- Xiongyan Wu
- Department of General SurgeryShanghai Key Laboratory of Gastric NeoplasmsShanghai Institute of Digestive SurgeryRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhijian Jin
- Department of General SurgeryShanghai Key Laboratory of Gastric NeoplasmsShanghai Institute of Digestive SurgeryRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Baolong Li
- Department of General SurgeryShanghai Key Laboratory of Gastric NeoplasmsShanghai Institute of Digestive SurgeryRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yifan Lu
- Department of General SurgeryShanghai Key Laboratory of Gastric NeoplasmsShanghai Institute of Digestive SurgeryRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Junyi Hou
- Department of General SurgeryShanghai Key Laboratory of Gastric NeoplasmsShanghai Institute of Digestive SurgeryRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lizhong Yao
- Department of General SurgeryShanghai Key Laboratory of Gastric NeoplasmsShanghai Institute of Digestive SurgeryRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhenjia Yu
- Department of General SurgeryShanghai Key Laboratory of Gastric NeoplasmsShanghai Institute of Digestive SurgeryRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qingqing Sang
- Department of General SurgeryShanghai Key Laboratory of Gastric NeoplasmsShanghai Institute of Digestive SurgeryRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Beiqin Yu
- Department of General SurgeryShanghai Key Laboratory of Gastric NeoplasmsShanghai Institute of Digestive SurgeryRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jianfang Li
- Department of General SurgeryShanghai Key Laboratory of Gastric NeoplasmsShanghai Institute of Digestive SurgeryRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Chen Li
- Department of General SurgeryShanghai Key Laboratory of Gastric NeoplasmsShanghai Institute of Digestive SurgeryRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Chao Yan
- Department of General SurgeryShanghai Key Laboratory of Gastric NeoplasmsShanghai Institute of Digestive SurgeryRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhenggang Zhu
- Department of General SurgeryShanghai Key Laboratory of Gastric NeoplasmsShanghai Institute of Digestive SurgeryRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Kaiwen Tang
- Department of General SurgeryShanghai Key Laboratory of Gastric NeoplasmsShanghai Institute of Digestive SurgeryRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bingya Liu
- Department of General SurgeryShanghai Key Laboratory of Gastric NeoplasmsShanghai Institute of Digestive SurgeryRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Liping Su
- Department of General SurgeryShanghai Key Laboratory of Gastric NeoplasmsShanghai Institute of Digestive SurgeryRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
33
|
Finger AM, Hendley AM, Figueroa D, Gonzalez H, Weaver VM. Tissue mechanics in tumor heterogeneity and aggression. Trends Cancer 2025:S2405-8033(25)00096-2. [PMID: 40307158 DOI: 10.1016/j.trecan.2025.04.004] [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: 08/27/2024] [Revised: 03/10/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025]
Abstract
Tumorigenesis ensues within a heterogeneous tissue microenvironment that promotes malignant transformation, metastasis and treatment resistance. A major feature of the tumor microenvironment is the heterogeneous population of cancer-associated fibroblasts and myeloid cells that stiffen the extracellular matrix. The heterogeneously stiffened extracellular matrix in turn activates cellular mechanotransduction and creates a hypoxic and metabolically hostile microenvironment. The stiffened extracellular matrix and elevated mechanosignaling also drive tumor aggression by fostering tumor cell growth, survival, and invasion, compromising antitumor immunity, expanding cancer stem cell frequency, and increasing mutational burden, which promote intratumor heterogeneity. Delineating the molecular mechanisms whereby tissue mechanics regulate these phenotypes should help to clarify the basis for tumor heterogeneity and cancer aggression and identify novel therapeutic targets that could improve patient outcome. Here, we discuss the role of the extracellular matrix in driving cancer aggression through its impact on tumor heterogeneity.
Collapse
Affiliation(s)
- Anna-Marie Finger
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA 94143; Current address: Liver Disease Research, Global Drug Discovery, Novo Nordisk A/S, Malov, Denmark
| | - Audrey Marie Hendley
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA 94143
| | - Diego Figueroa
- Department of Radiation Oncology, Department of Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Hugo Gonzalez
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA 94143; Current address: Laboratory of Tumor Microenvironment and Metastasis, Centro Ciencia & Vida, Santiago, Chile
| | - Valerie Marie Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA 94143; Department of Radiation Oncology, Department of Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
34
|
Moomin A, Gawusu S, Sidik AI, Tizumah MW, Adam MK, Adu-Gyamfi PKT, Mensah KB. Dietary Fiber and Cancer Management: A Twenty-Five-Year Bibliometric Review of Research Trends and Directions. BIOMED RESEARCH INTERNATIONAL 2025; 2025:5086946. [PMID: 40330094 PMCID: PMC12055321 DOI: 10.1155/bmri/5086946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 03/28/2025] [Indexed: 05/08/2025]
Abstract
Background: The global burden of cancer necessitates innovative approaches to its management and treatment. Traditional treatments like radiotherapy, immunotherapy, surgery, and chemotherapy, carry significant side effects that impact patient quality of life. Dietary fiber has attracted research interest as a potential mitigator of cancer progression and a supportive agent in cancer treatment. This bibliometric study analyzes trends in research connecting dietary fiber, cancer therapy, and gut health from April 1999 to May 2024. Methods and Results: Web of Science, PubMed, and Scopus databases were used to retrieve peer-reviewed publications from April 1999 to May 2024 on dietary fiber and cancer management. The study identifies a rising scholarly interest in dietary fiber's role in cancer management, focusing significantly on the interactions between dietary fiber and gut microbiota. These interactions are particularly noted for their influence on inflammation and cancer metastasis. The study highlights evolving research themes, the importance of specific fiber types in cancer progression, and highlights persistent foundational themes like glycosylation. Emerging areas include personalized nutrition and innovative therapeutic approaches. The geographical and institutional contributions, mainly from the United States and China, underline the significance of collaborative and interdisciplinary efforts in advancing research. Conclusion: This analysis emphasizes integrating dietary strategies in comprehensive cancer care and aims to address research gaps to develop more effective and patient-centered cancer therapy and prevention strategies.
Collapse
Affiliation(s)
- Aliu Moomin
- Rowett Institute, University of Aberdeen, Aberdeen, Scotland, UK
- Aberdeen Cancer Centre, University of Aberdeen, Aberdeen, Scotland, UK
| | - Sidique Gawusu
- Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Marizuk Waris Tizumah
- Department of Medical laboratory/Medical Imaging Technology, Accra Technical University, Accra, Greater Accra, Ghana
- Medical Centre, University of Ghana, Accra, Greater Accra, Ghana
| | - Maridia Kunateh Adam
- Department of Data Science, University of East London, London, England, UK
- Department of Dietetics, Robert Gordon University, Aberdeen, Scotland, UK
| | | | - Kwesi Boadu Mensah
- Department of Pharmacology, Kwame Nkrumah University of Science and Technology, Kumasi, Ashanti Region, Ghana
| |
Collapse
|
35
|
Timilsina S, Huang JY, Abdelfattah N, Medina D, Singh D, Abdulsahib S, Subbarayalu P, Do TP, Venkata PP, Nirzhor S, Prochnau J, Bhandari M, Zheng S, Chen Y, Huang G, Mukherjee N, Hromas R, Sung P, Kaklamani V, Vadlamudi R, Zhang N, Rao MK. Epigenetic silencing of DNA sensing pathway by FOXM1 blocks stress ligand-dependent antitumor immunity and immune memory. Nat Commun 2025; 16:3967. [PMID: 40295473 PMCID: PMC12037779 DOI: 10.1038/s41467-025-59186-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 04/14/2025] [Indexed: 04/30/2025] Open
Abstract
The interplay between tumor cells and the microenvironment significantly influences cancer progression. Here, we report a significant role of the transcription factor FOXM1 in shaping the tumor immune landscape. Single-cell sequencing reveals that tumor-intrinsic FOXM1 creates an immune-suppressive tumor microenvironment by inhibiting expression of stress ligands (including ULBP1) on cancer cells, thereby blocking NKG2D-NKG2DL interactions critical for priming natural killer- and T cell-mediated cytotoxicity of cancer cells. FOXM1 suppresses ULBP1 expression by epigenetically silencing the DNA-sensing protein STING using a DNMT1-UHRF1 complex, which in turn inhibits the unfolded protein response protein CHOP from activating ULBP1. Importantly, cancer patients with higher levels of FOXM1 and DNMT1, and lower levels of STING and ULBP1, have worse survival and are less responsive to immunotherapy. Collectively, our findings provide key insight into how a tumor-intrinsic transcription factor epigenetically shapes the tumor immune microenvironment, with strong implications for refining existing and designing new cancer immunotherapies.
Collapse
Affiliation(s)
| | - Jian Yu Huang
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
| | - Nourhan Abdelfattah
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, USA
| | - Daisy Medina
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Deepika Singh
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Shahad Abdulsahib
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Panneerdoss Subbarayalu
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Trong Phat Do
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Prabhakar Pitta Venkata
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Saif Nirzhor
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Jack Prochnau
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
| | - Mukund Bhandari
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Siyuan Zheng
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Population Health Sciences, UT Health San Antonio, San Antonio, TX, USA
| | - Yidong Chen
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Population Health Sciences, UT Health San Antonio, San Antonio, TX, USA
| | - Gang Huang
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | | | - Robert Hromas
- Department of Medicine, UT Health, San Antonio, TX, USA
| | - Patrick Sung
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Biochemistry & Structural Biology, UT Health San Antonio, San Antonio, TX, USA
| | | | - Ratna Vadlamudi
- Department of Obstetrics and Gynecology, UT Health San Antonio, San Antonio, TX, USA
- Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX, USA
| | - Nu Zhang
- Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX, USA
- Department of Microbiology, Immunology & Molecular Genetics, UT Health, San Antonio, TX, USA
| | - Manjeet K Rao
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA.
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, USA.
| |
Collapse
|
36
|
Naffaa MM, Al-Ewaidat OA, Gogia S, Begiashvili V. Neoantigen-based immunotherapy: advancing precision medicine in cancer and glioblastoma treatment through discovery and innovation. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002313. [PMID: 40309350 PMCID: PMC12040680 DOI: 10.37349/etat.2025.1002313] [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: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
Neoantigen-based immunotherapy has emerged as a transformative approach in cancer treatment, offering precision medicine strategies that target tumor-specific antigens derived from genetic, transcriptomic, and proteomic alterations unique to cancer cells. These neoantigens serve as highly specific targets for personalized therapies, promising more effective and tailored treatments. The aim of this article is to explore the advances in neoantigen-based therapies, highlighting successful treatments such as vaccines, tumor-infiltrating lymphocyte (TIL) therapy, T-cell receptor-engineered T cells therapy (TCR-T), and chimeric antigen receptor T cells therapy (CAR-T), particularly in cancer types like glioblastoma (GBM). Advances in technologies such as next-generation sequencing, RNA-based platforms, and CRISPR gene editing have accelerated the identification and validation of neoantigens, moving them closer to clinical application. Despite promising results, challenges such as tumor heterogeneity, immune evasion, and resistance mechanisms persist. The integration of AI-driven tools and multi-omic data has refined neoantigen discovery, while combination therapies are being developed to address issues like immune suppression and scalability. Additionally, the article discusses the ongoing development of personalized immunotherapies targeting tumor mutations, emphasizing the need for continued collaboration between computational and experimental approaches. Ultimately, the integration of cutting-edge technologies in neoantigen research holds the potential to revolutionize cancer care, offering hope for more effective and targeted treatments.
Collapse
Affiliation(s)
- Moawiah M Naffaa
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ola A Al-Ewaidat
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Sopiko Gogia
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Valiko Begiashvili
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| |
Collapse
|
37
|
Tan X, He W, Chen T, Yang W, Huang D, Zhang H, Luo Y, Lu M, Zhang Z, Ji J, Liu H. Immune-related gene expression with colorectal cancer risk: a Mendelian randomization analysis. Discov Oncol 2025; 16:617. [PMID: 40285908 PMCID: PMC12033153 DOI: 10.1007/s12672-025-02291-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/01/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND The potential role of immune cells and the precise contributions of genes from various immune cells in the development of colorectal cancer (CRC) remains unclear. We aimed to investigate the potential causal relationships between specific immune cell genes and the development of CRC by performing Mendelian randomization (MR) analysis. METHODS The cis-expression quantitative trait loci (eQTLs) data for type-specific immune cells were used to perform MR analysis. Outcomes data were obtained from the latest Genome Wide Association Studies (GWAS), comprising 78,473 European ancestry CRC cases and 107,143 controls. A few sensitivity analyses, encompassing pleiotropy and colocalization analyses, along with the application of False Discovery Rate (FDR) correction, were carried out to mitigate potential biases. RESULTS A total of 395 genes were found to be associated with CRC. After FDR correction, a total of 47 genes across 14 immune cell types were identified. Notably, the FHL3 gene showed the strongest association with CRC in multiple immune cell types, including CD4 + naïve and central memory T cells (CD4NC), CD4 + T cells with an effector memory or central memory phenotype (CD4ET), CD8 + naïve and central memory T cells (CD8NC), CD8 + T cells with an effector memory phenotype (CD8ET), and NK recruiting cells (NKR). Sensitivity analyses and pleiotropy assessments excluded the bias due to weak instruments and linkage disequilibrium. Additionally, the associations were supported by strong evidence of colocalization with CRC. The expression of FHL3 in normal tissue was higher than that in the tumor tissue at the mRNA expression and single-cell levels, and low mRNA expression of FHL3 was associated with shorter survival time in CRC patients. CONCLUSIONS Our study provides a novel perspective on the potential causal link between the low expression of FHL3 gene and the risk of CRC. More evidence is essential to further reveal the biological mechanisms underlying this association.
Collapse
Affiliation(s)
- Xin Tan
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenjun He
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tao Chen
- Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Research Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weihao Yang
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Donghua Huang
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hengyi Zhang
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yangyang Luo
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengting Lu
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhenzhan Zhang
- Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Jianguang Ji
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao, Macao SAR, China.
| | - Hao Liu
- Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
38
|
Wang J, Tao X, Zhu J, Dai Z, Du Y, Xie Y, Chu X, Fu G, Lei Z. Tumor organoid-immune co-culture models: exploring a new perspective of tumor immunity. Cell Death Discov 2025; 11:195. [PMID: 40268893 PMCID: PMC12019369 DOI: 10.1038/s41420-025-02407-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 04/25/2025] Open
Abstract
Recent advancements in technology have significantly expanded the scope of tumor research, progressing from the study of individual cells to more intricate tissue and organ-level analyses. Tumor organoids have emerged as a highly realistic platform for investigating tumor growth, development, and their interactions with the surrounding microenvironment. However, a notable limitation of these organoids is their lack of the diverse cellular composition typically observed in actual tumors, which hinders their ability to fully replicate the complexity of the tumor microenvironment. Immune cells play a pivotal role, and tumor immunology has become a major research hotspot. Research in tumor immunology aims to elucidate how the immune system recognizes and attacks tumor cells, as well as how tumor cells evade immune surveillance. In recent years, there has been growing interest in co-culturing immune cells with tumor organoids, an approach that has yielded valuable insights into the intricate interactions between tumors and the immune system. The aim of this paper is to review and discuss the progress achieved in co-culturing tumor organoids with immune cells. By doing so, we hope to offer a new perspective and enhance our understanding of the complexity and diversity inherent in the tumor microenvironment.
Collapse
Affiliation(s)
- Jing Wang
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Xiaoyue Tao
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jialong Zhu
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhe Dai
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuanyang Du
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yiyang Xie
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoyuan Chu
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China.
| | - Gongbo Fu
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China.
| | - Zengjie Lei
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China.
| |
Collapse
|
39
|
Armah J, Alzahid SK, Pei Q, Stacciarini JMR, Heldermon C, Starkweather A. Exercise to Manage Fatigue During and After Chemotherapy in Adolescents and Young Adults With Cancer: A Systematic Review and Meta-Analysis. Oncol Nurs Forum 2025; 52:E77-E92. [PMID: 40293930 PMCID: PMC12056843 DOI: 10.1188/25.onf.e77-e92] [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: 07/01/2024] [Accepted: 10/02/2024] [Indexed: 04/30/2025]
Abstract
PROBLEM IDENTIFICATION There is a gap in knowledge about the efficacy of exercise in managing cancer-related fatigue (CRF) in adolescents and young adults (AYAs) during and after chemotherapy. LITERATURE SEARCH A systematic search was conducted in Scopus®, PubMed®, and CINAHL®, as well as citation searching, for studies about the impact of exercise on CRF in the AYA population. Abstract and full-text screening of 2,234 studies produced 15 studies for systematic review and 13 for meta-analysis. DATA EVALUATION Effect size was calculated using standardized mean difference and confidence intervals. Subgroup analysis was conducted to assess the impact of various exercise types on CRF. A meta-regression was performed using exercise frequency, intensity, and duration as predictors to determine relationships with effect size on CRF and adherence to exercise. SYNTHESIS Exercise had a large effect size on CRF in AYAs. No difference in effect size estimates for exercise type subgroups was observed. A linear association was found between exercise duration and CRF (p = 0.005), and exercise intensity and adherence (p = 0.037). IMPLICATIONS FOR PRACTICE Exercise is effective in managing CRF among AYAs, and efforts should be aimed toward including it in routine oncologic care.
Collapse
|
40
|
Deslandes B, Wu X, Lee MA, Goudswaard LJ, Jones GW, Gsur A, Lindblom A, Ogino S, Vymetalkova V, Wolk A, Wu AH, Huyghe JR, Peters U, Phipps AI, Thomas CE, Pai RK, Grant RC, Buchanan DD, Yarmolinsky J, Gunter MJ, Zheng J, Hazelwood E, Vincent EE. Transcriptome-wide Mendelian randomisation exploring dynamic CD4+ T cell gene expression in colorectal cancer development. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.15.25325863. [PMID: 40321251 PMCID: PMC12047913 DOI: 10.1101/2025.04.15.25325863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Background Recent research has identified a potential protective effect of higher numbers of circulating lymphocytes on colorectal cancer (CRC) development. However, the importance of different lymphocyte subtypes and activation states in CRC development and the biological pathways driving this relationship remain poorly understood and warrant further investigation. Specifically, CD4+ T cells - a highly dynamic lymphocyte subtype - undergo remodelling upon activation to induce the expression of genes critical for their effector function. Previous studies investigating their role in CRC risk have used bulk tissue, limiting our current understanding of the role of these cells to static, non-dynamic relationships only. Methods Here, we combined two genetic epidemiological methods - Mendelian randomisation (MR) and genetic colocalisation - to evaluate evidence for causal relationships of gene expression on CRC risk across multiple CD4+ T cell subtypes and activation stage. Genetic proxies were obtained from single-cell transcriptomic data, allowing us to investigate the causal effect of expression of 1,805 genes across five CD4+ T cell activation states on CRC risk (78,473 cases; 107,143 controls). We repeated analyses stratified by CRC anatomical subsites and sex, and performed a sensitivity analysis to evaluate whether the observed effect estimates were likely to be CD4+ T cell-specific. Results We identified six genes with evidence (FDR-P<0.05 in MR analyses and H4>0.8 in genetic colocalisation analyses) for a causal role of CD4+ T cell expression in CRC development - FADS2, FHL3, HLA-DRB1, HLA-DRB5, RPL28, and TMEM258. We observed differences in causal estimates of gene expression on CRC risk across different CD4+ T cell subtypes and activation timepoints, as well as CRC anatomical subsites and sex. However, our sensitivity analysis revealed that the genetic proxies used to instrument gene expression in CD4+ T cells also act as eQTLs in other tissues, highlighting the challenges of using genetic proxies to instrument tissue-specific expression changes. Conclusions Our study demonstrates the importance of capturing the dynamic nature of CD4+ T cells in understanding disease risk, and prioritises genes for further investigation in cancer prevention research.
Collapse
Affiliation(s)
- Benedita Deslandes
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Xueyan Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Matthew A. Lee
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, WHO, Lyon, France
| | - Lucy J. Goudswaard
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Gareth W. Jones
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Andrea Gsur
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Annika Lindblom
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Shuji Ogino
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Veronika Vymetalkova
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Alicja Wolk
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna H. Wu
- University of Southern California, Department of Population and Public Health Sciences, Los Angeles, California, USA
| | - Jeroen R. Huyghe
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Amanda I. Phipps
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Claire E. Thomas
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Rish K. Pai
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Robert C Grant
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Daniel D. Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, VIC 3010, Australia
- University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC 3010, Australia
- Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, Melbourne, VIC 3000, Australia
| | - James Yarmolinsky
- Cancer Epidemiology and Prevention Research Unit, School of Public Health, Imperial College London, London, United Kingdom
| | - Marc J. Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, WHO, Lyon, France
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Emma Hazelwood
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Emma E. Vincent
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
41
|
Chung MW, Tzeng CC, Huang YC, Wei KC, Hsu PW, Chuang CC, Lin YJ, Chen KT, Lee CC. Neutrophil-to-lymphocyte ratio dynamics: prognostic value and potential for surveilling glioblastoma recurrence. BMC Cancer 2025; 25:709. [PMID: 40241016 PMCID: PMC12004828 DOI: 10.1186/s12885-025-14118-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025] Open
Abstract
PURPOSE Glioblastoma (GBM) is a challenging malignancy with a poor prognosis. While the neutrophil-to-lymphocyte ratio (NLR) is reported to correlate with the prognosis, the significance of changes in the NLR and its prognostic value in GBM remain unclear. This study aims to evaluate changes in the NLR and its predictive value for GBM prognosis and recurrence. METHODS The cohort included 69 newly-diagnosed GBM patients undergoing a standard treatment protocol. NLR was assessed at multiple time points. The dynamic change in NLR (dNLR), defined as the NLR at the point of interest (post-CCRT or post-Stupp) divided by the preoperative NLR, also was assessed. Univariate and multivariate COX regression analyses were conducted to assess the association between the NLR, dNLR and overall survival (OS) and progression-free survival (PFS). RESULTS Univariate analysis revealed that age at diagnosis ≥ 70 (p = 0.019) and post-Stupp dNLR ≥ 1.3 (p = 0.006) were significantly associated with shorter OS. Significant correlations were found between pre-operative KPS ≥ 60 (p = 0.017), gross total resection (p = 0.042), post-Stupp dNLR ≥ 1.3 (p = 0.043) and PFS. Multivariate analysis showed age at diagnosis ≥ 70, pre-operative KPS ≥ 60, post-Stupp NLR ≥ 5 and dNLR ≥ 1.3 were significantly associated with a shorter OS. Significant correlation was found between pre-operative KPS ≥ 60 and PFS. CONCLUSION This study revealed that post-Stupp NLR ≥ 5 and dNLR ≥ 1.3 correlated significantly with a worse glioblastoma prognosis in OS, and dNLR might be more reliable. These two parameters are potentially surveilling markers for glioblastoma recurrence, however further studies are warranted.
Collapse
Affiliation(s)
- Meng-Wu Chung
- Department of Neurosurgery, Chang Gung Memorial Hospital, 5 Fuxing St., Guishan Dist, 33305, Taoyuan, Taiwan
| | - Ching-Chieh Tzeng
- Department of Medical Education, Chang Gung Memorial Hospital, 33305, Taoyuan, Taiwan
| | - Yin-Cheng Huang
- Department of Neurosurgery, Chang Gung Memorial Hospital, 5 Fuxing St., Guishan Dist, 33305, Taoyuan, Taiwan
- Chang Gung University, 33302, Taoyuan, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, Chang Gung Memorial Hospital, 5 Fuxing St., Guishan Dist, 33305, Taoyuan, Taiwan
- Chang Gung University, 33302, Taoyuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, 33305, Taoyuan, Taiwan
- Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, 236017, New Taipei, Taiwan
| | - Peng-Wei Hsu
- Department of Neurosurgery, Chang Gung Memorial Hospital, 5 Fuxing St., Guishan Dist, 33305, Taoyuan, Taiwan
- School of Medicine, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Chi-Cheng Chuang
- Department of Neurosurgery, Chang Gung Memorial Hospital, 5 Fuxing St., Guishan Dist, 33305, Taoyuan, Taiwan
- Chang Gung University, 33302, Taoyuan, Taiwan
| | - Ya-Jui Lin
- Department of Neurosurgery, Chang Gung Memorial Hospital, 5 Fuxing St., Guishan Dist, 33305, Taoyuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, 33305, Taoyuan, Taiwan
- Division of Natural Product, Graduate Institute of Biomedical Sciences, Chang Gung University, 33302, Taoyuan, Taiwan
| | - Ko-Ting Chen
- Department of Neurosurgery, Chang Gung Memorial Hospital, 5 Fuxing St., Guishan Dist, 33305, Taoyuan, Taiwan
- Chang Gung University, 33302, Taoyuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, 33305, Taoyuan, Taiwan
| | - Cheng-Chi Lee
- Department of Neurosurgery, Chang Gung Memorial Hospital, 5 Fuxing St., Guishan Dist, 33305, Taoyuan, Taiwan.
- Chang Gung University, 33302, Taoyuan, Taiwan.
| |
Collapse
|
42
|
Quan X, Deng Y, Liu Z, Gao Z, Yi H, Li M. LASSO-derived nomogram prediction model for lymph node metastasis in colorectal cancer: a retrospective analysis. PeerJ 2025; 13:e19148. [PMID: 40247833 PMCID: PMC12005177 DOI: 10.7717/peerj.19148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 02/19/2025] [Indexed: 04/19/2025] Open
Abstract
Objective This study aims to develop a prediction model for lymph node metastasis (LNM) in colorectal cancer (CRC) patients using common clinicopathologic data and a nomogram. The model seeks to uncover correlations between LNM and clinical indicators, providing an effective tool to identify high-risk patients, aiding clinical decision-making, and enhancing patient prognosis. Methods We conducted a retrospective analysis of CRC patients diagnosed between January 2021 and December 2023 at Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University. Risk predictors for LNM were identified through comparative analysis and Least Absolute Shrinkage and Selection Operator (LASSO) logistic regression. Nomograms were then utilized to predict the probability of metastasis, and their performance was assessed using calibration curves, receiver operating characteristic (ROC) curves, and decision curve analysis. Results The study comprised 869 CRC patients, with 435 cases allocated to the training set and 434 cases to the validation set. First, 12 potential risk factors for LNM in CRC patients were identified through comparative analysis in the training set. Next, nine independent predictors (T stage, vascular tumor thrombus, PMS2, MSH2, KRAS, BRAF, PIK3CA, leukocyte, and neutrophil) of LNM occurrence were refined using LASSO regression and multivariate logistic regression models. Subsequently, a clinical nomogram was developed based on these independent predictors of LNM. The nomogram exhibited a C-index of 0.751 (95% CI [0.728-0.774]), indicating its robust predictive value, which was further validated in the independent validation set. Conclusion T stage, vascular tumor thrombus, PMS2, MSH2, KRAS, BRAF, and neutrophil emerged as significant risk factors for LNM in CRC, while leukocytes appeared to be protective. These findings emphasize the importance of comprehensive risk assessment and personalized therapeutic strategies in CRC management.
Collapse
Affiliation(s)
- Xiyun Quan
- Department of Pathology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Yi Deng
- Department of Pathology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Zhimin Liu
- Neurosurgery, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Zhenqin Gao
- Department of Pathology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Huimei Yi
- Department of Pathology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Ming Li
- Health Management Center, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| |
Collapse
|
43
|
Rodriguez-Sevilla JJ, Ganan-Gomez I, Kumar B, Thongon N, Ma F, Chien KS, Kim YJ, Yang H, Loghavi S, Tan R, Adema V, Li Z, Tanaka T, Uryu H, Kanagal-Shamanna R, Al-Atrash G, Bejar R, Banerjee PP, Lynn Cha S, Montalban-Bravo G, Dougherty M, Fernandez Laurita MC, Wheeler N, Jia B, Papapetrou EP, Izzo F, Dueñas DE, McAllen S, Gu Y, Todisco G, Ficara F, Della Porta MG, Jain A, Takahashi K, Clise-Dwyer K, Halene S, Bertilaccio MTS, Garcia-Manero G, Daher M, Colla S. Natural killer cells' functional impairment drives the immune escape of pre-malignant clones in early-stage myelodysplastic syndromes. Nat Commun 2025; 16:3450. [PMID: 40216768 PMCID: PMC11992119 DOI: 10.1038/s41467-025-58662-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Dissecting the preneoplastic disease states' biological mechanisms that precede tumorigenesis can lead to interventions that can slow down disease progression and/or mitigate disease-related comorbidities. Myelodysplastic syndromes (MDS) cannot be cured by currently available pharmacological therapies, which fail to eradicate aberrant hematopoietic stem cells (HSCs), most of which are mutated by the time of diagnosis. Here, we sought to elucidate how MDS HSCs evade immune surveillance and expand in patients with clonal cytopenias of undetermined significance (CCUS), the pre-malignant stage of MDS. We used multi-omic single-cell approaches and functional in vitro studies to show that immune escape at disease initiation is mainly mediated by mutant, dysfunctional natural killer (NK) cells with impaired cytotoxic capability against cancer cells. Preclinical in vivo studies demonstrated that injecting NK cells from healthy donors efficiently depleted CCUS mutant cells while allowing normal cells to regenerate hematopoiesis. Our findings suggest that early intervention with adoptive cell therapy can prevent or delay the development of MDS.
Collapse
Affiliation(s)
| | - Irene Ganan-Gomez
- Department of Leukemia, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Bijender Kumar
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Natthakan Thongon
- Department of Leukemia, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Feiyang Ma
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Kelly S Chien
- Department of Leukemia, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Yi J Kim
- Department of Genomic Medicine, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Hui Yang
- Department of Leukemia, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Sanam Loghavi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roselyn Tan
- Moores Cancer Center, University of California San Diego, Moores Cancer Center, San Diego, CA, USA
| | - Vera Adema
- Department of Leukemia, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Zongrui Li
- Department of Genomic Medicine, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Tomoyuki Tanaka
- Department of Genomic Medicine, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Hidetaka Uryu
- Department of Genomic Medicine, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gheath Al-Atrash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rafael Bejar
- Moores Cancer Center, University of California San Diego, Moores Cancer Center, San Diego, CA, USA
| | - Pinaki Prosad Banerjee
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sophia Lynn Cha
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Max Dougherty
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria Claudina Fernandez Laurita
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Noelle Wheeler
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Baosen Jia
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eirini P Papapetrou
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Franco Izzo
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniela E Dueñas
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Salome McAllen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yiqian Gu
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Gabriele Todisco
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
| | - Francesca Ficara
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
- Istituto di Ricerca Genetica e Biomedica, National Research Council, 20090, Milan, Italy
| | - Matteo Giovanni Della Porta
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
| | - Abhinav Jain
- Department of Epigenetics and Molecular Carcinogenesis, Center for Cancer Epigenetics, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Koichi Takahashi
- Department of Leukemia, The University of MD Anderson Cancer Center, Houston, TX, USA
- Department of Genomic Medicine, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Karen Clise-Dwyer
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | | | | | - May Daher
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Simona Colla
- Department of Leukemia, The University of MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
44
|
Wachter E, Fox LH, Lu Z, Jones AD, Casto ND, Waltz SE. RON Receptor Signaling and the Tumor Microenvironment. Genes (Basel) 2025; 16:437. [PMID: 40282397 PMCID: PMC12026484 DOI: 10.3390/genes16040437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
The immune microenvironment plays a critical role in tumor growth and development. Immune activation within the tumor microenvironment is dynamic and can be modulated by tumor intrinsic and extrinsic signaling. The RON receptor tyrosine kinase is canonically associated with growth signaling and wound healing, and this receptor is frequently overexpressed in a variety of cancers. Epithelial cells, macrophages, dendritic cells, and fibroblasts express RON, presenting an important axis by which RON overexpressing tumors influence the tumor microenvironment. This review synthesizes the existing literature on the roles of tumor cell-intrinsic and -extrinsic RON signaling, highlighting areas of interest and gaps in knowledge that show potential for future studies.
Collapse
Affiliation(s)
- Emily Wachter
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Levi H. Fox
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Zhixin Lu
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Angelle D. Jones
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Nicholas D. Casto
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Susan E. Waltz
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
- Research Service, Cincinnati Veterans Affairs Hospital Medical Center, Cincinnati, OH 45220, USA
| |
Collapse
|
45
|
Wahnou H, El Kebbaj R, Hba S, Ouadghiri Z, El Faqer O, Pinon A, Liagre B, Limami Y, Duval RE. Neutrophils and Neutrophil-Based Drug Delivery Systems in Anti-Cancer Therapy. Cancers (Basel) 2025; 17:1232. [PMID: 40227814 PMCID: PMC11988188 DOI: 10.3390/cancers17071232] [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: 03/08/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/15/2025] Open
Abstract
Neutrophils, the most abundant white blood cells, play a dual role in cancer progression. While they can promote tumor growth, metastasis, and immune suppression, they also exhibit anti-tumorigenic properties by attacking cancer cells and enhancing immune responses. This review explores the complex interplay between neutrophils and the tumor microenvironment (TME), highlighting their ability to switch between pro- and anti-tumor phenotypes based on external stimuli. Pro-tumorigenic neutrophils facilitate tumor growth through mechanisms such as neutrophil extracellular traps (NETs), secretion of pro-inflammatory cytokines, and immune evasion strategies. They contribute to angiogenesis, tumor invasion, and metastasis by releasing vascular endothelial growth factor (VEGF) and matrix metalloproteinases (MMPs). Conversely, anti-tumor neutrophils enhance cytotoxicity by generating reactive oxygen species (ROS), promoting antibody-dependent cell-mediated cytotoxicity (ADCC), and activating other immune cells such as cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells. Recent advances in neutrophil-based drug delivery systems have harnessed their tumor-homing capabilities to improve targeted therapy. Neutrophil-mimicking nanoparticles and membrane-coated drug carriers offer enhanced drug accumulation in tumors, reduced systemic toxicity, and improved therapeutic outcomes. Additionally, strategies to modulate neutrophil activity, such as inhibiting their immunosuppressive functions or reprogramming them towards an anti-tumor phenotype, are emerging as promising approaches in cancer immunotherapy. Understanding neutrophil plasticity and their interactions with the TME provides new avenues for therapeutic interventions. Targeting neutrophil-mediated mechanisms could enhance existing cancer treatments and lead to the development of novel immunotherapies, ultimately improving patient survival and clinical outcomes.
Collapse
Affiliation(s)
- Hicham Wahnou
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
| | - Riad El Kebbaj
- Sciences and Engineering of Biomedicals, Biophysics and Health Laboratory, Higher Institute of Health Sciences, Hassan First University, Settat 26000, Morocco;
| | - Soufyane Hba
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (A.P.); (B.L.)
| | - Zaynab Ouadghiri
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
| | - Othman El Faqer
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
| | - Aline Pinon
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (A.P.); (B.L.)
| | - Bertrand Liagre
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (A.P.); (B.L.)
| | - Youness Limami
- Sciences and Engineering of Biomedicals, Biophysics and Health Laboratory, Higher Institute of Health Sciences, Hassan First University, Settat 26000, Morocco;
| | | |
Collapse
|
46
|
Mongiovi JM, Townsend MK, Vitonis AF, Harris HR, Doherty JA, Babic A, Hecht JL, Soong TR, Titus L, Conejo-Garcia JR, Fridley BL, Tworoger SS, Terry KL, Sasamoto N. Associations between Parity, History of Breastfeeding, and T-cell Profile of Ovarian Tumors. Cancer Epidemiol Biomarkers Prev 2025; 34:550-559. [PMID: 39912719 PMCID: PMC11968234 DOI: 10.1158/1055-9965.epi-24-1414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/13/2024] [Accepted: 02/04/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Parity and breastfeeding are associated with systemic changes in maternal inflammation and reduced risk of ovarian cancer, but little is known about their impact on the ovarian tumor immune microenvironment. METHODS We evaluated the associations of self-reported parity and history of breastfeeding with tumor-infiltrating T cells among 1,706 ovarian carcinoma cases with tumor tissue collected across four studies. The abundance of tumor-infiltrating T cells was measured by multiplex immunofluorescence in tumor tissue microarrays. ORs and 95% confidence intervals (CI) for the positivity of tumor immune cells were calculated using beta-binomial models and stratified by histotype. RESULTS Compared with ovarian tumors in nulliparous women, there was no association between parity and ovarian tumor T-cell abundance among all histotypes combined but suggestion of increased cytotoxic T cells and T-cell exhaustion among parous women with clear-cell tumors. When restricted to parous women, history of breastfeeding was associated with increased odds for all T-cell types [i.e., total T, cytotoxic T, helper T (Th), regulatory T, and exhausted T cells], with ORs ranging from 1.11 to 1.42. For every 6 months of breastfeeding, we observed increased odds of activated Th-cell infiltration (CD3+CD4+CD69+; OR, 1.13, 95% CI, 0.99-1.29), with a similar association for high-grade serous tumors, but lower odds in clear-cell tumors (OR, 0.43, 95% CI, 0.21-0.87). CONCLUSIONS History of breastfeeding may alter the ovarian tumor immune microenvironment by modulating the abundance of tumor-infiltrating T cells. IMPACT Although replication is required, history of breastfeeding may play a role in the activation of the ovarian tumor immune response.
Collapse
Affiliation(s)
- Jennifer M Mongiovi
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Mary K Townsend
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University School of Medicine, Portland, Oregon
| | - Allison F Vitonis
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Holly R Harris
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, Washington
- School of Public Health, University of Washington, Seattle, Washington
| | - Jennifer A Doherty
- Department of Population Health Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Ana Babic
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Division of Medical Oncology, Dana Farber Cancer Center, Boston, Massachusetts
| | - Jonathan L Hecht
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - T Rinda Soong
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Linda Titus
- Department of Epidemiology, Dartmouth Geisel School of Medicine, Hanover, New Hampshire
| | - Jose R Conejo-Garcia
- Department of Integrative Immunobiology, Duke School of Medicine, Durham, North Carolina
| | - Brooke L Fridley
- Division of Health Services and Outcomes Research, Children's Mercy Kansas City, Kansas City, Missouri
| | - Shelley S Tworoger
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University School of Medicine, Portland, Oregon
| | - Kathryn L Terry
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Naoko Sasamoto
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
47
|
Bhattacharya R, Avdieiev SS, Bukkuri A, Whelan CJ, Gatenby RA, Tsai KY, Brown JS. The Hallmarks of Cancer as Eco-Evolutionary Processes. Cancer Discov 2025; 15:685-701. [PMID: 40170539 DOI: 10.1158/2159-8290.cd-24-0861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/19/2024] [Accepted: 01/28/2025] [Indexed: 04/03/2025]
Abstract
SIGNIFICANCE Viewing the hallmarks as a sequence of adaptations captures the "why" behind the "how" of the molecular changes driving cancer. This eco-evolutionary view distils the complexity of cancer progression into logical steps, providing a framework for understanding all existing and emerging hallmarks of cancer and developing therapeutic interventions.
Collapse
Affiliation(s)
- Ranjini Bhattacharya
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Cancer Biology, University of South Florida, Tampa, Florida
| | - Stanislav S Avdieiev
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Anuraag Bukkuri
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Center for Evolutionary Biology and Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Christopher J Whelan
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Robert A Gatenby
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kenneth Y Tsai
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Tumor Microenvironment & Metastasis, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Joel S Brown
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
48
|
Chicón‐Bosch M, Sánchez‐Serra S, Rosàs‐Lapeña M, Costa‐Fraga N, Besalú‐Velázquez J, Illa‐Bernadí J, Mateo‐Lozano S, Cidre‐Aranaz F, Grünewald TG, Díaz‐Lagares Á, Lopez‐Alemany R, Tirado ÒM. Multi-omics profiling reveals key factors involved in Ewing sarcoma metastasis. Mol Oncol 2025; 19:1002-1028. [PMID: 39757762 PMCID: PMC11977646 DOI: 10.1002/1878-0261.13788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/28/2024] [Accepted: 11/14/2024] [Indexed: 01/07/2025] Open
Abstract
Ewing sarcoma (EWS) is the second most common bone tumor affecting children and young adults, with dismal outcomes for patients with metastasis at diagnosis. Mechanisms leading to metastasis remain poorly understood. To deepen our knowledge on EWS progression, we have profiled tumors and metastases from a spontaneous metastasis mouse model using a multi-omics approach. Combining transcriptomics, proteomics, and methylomics analyses, we identified signaling cascades and candidate genes enriched in metastases that could be modulating aggressiveness in EWS. Phenotypical validation of two of these candidates, cyclic AMP-responsive element-binding protein 1 (CREB1) and lipoxygenase homology domain-containing protein 1 (LOXHD1), showed an association with migration and clonogenic abilities. Moreover, previously described CREB1 downstream targets were present amongst the metastatic-enriched results. Combining the different omics datasets, we identified FYVE, RhoGEF, and PH domain-containing protein 4 (FGD4) as a CREB1 target interconnecting the different EWS biological layers (RNA, protein and methylation status) and whose high expression is associated with worse clinical outcome. Further studies will provide insight into EWS metastasis mechanisms and ultimately improve survival rates for EWS patients.
Collapse
Affiliation(s)
- Mariona Chicón‐Bosch
- Sarcoma Research GroupInstitut d'Investigació Biomèdica de Bellvitge (IDIBELL), Oncobell, L'Hospitalet de LlobregatBarcelonaSpain
| | - Sara Sánchez‐Serra
- Sarcoma Research GroupInstitut d'Investigació Biomèdica de Bellvitge (IDIBELL), Oncobell, L'Hospitalet de LlobregatBarcelonaSpain
- Universitat de Barcelona (UB)BarcelonaSpain
| | - Marta Rosàs‐Lapeña
- Sarcoma Research GroupInstitut d'Investigació Biomèdica de Bellvitge (IDIBELL), Oncobell, L'Hospitalet de LlobregatBarcelonaSpain
| | - Nicolás Costa‐Fraga
- Epigenomics Unit, Cancer Epigenomics, Translational Medical Oncology (ONCOMET), Health Research Institute of Santiago (IDIS)University Clinical Hospital of Santiago (CHUS/SERGAS)Santiago de CompostelaSpain
- Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry SchoolUniversidade de Santiago de Compostela (USC)Spain
- Universidad de Santiago de Compostela (USC)Spain
- Department of Clinical AnalysisUniversity Hospital Complex of Santiago de Compostela (CHUS)Spain
| | - Judit Besalú‐Velázquez
- Sarcoma Research GroupInstitut d'Investigació Biomèdica de Bellvitge (IDIBELL), Oncobell, L'Hospitalet de LlobregatBarcelonaSpain
| | - Janet Illa‐Bernadí
- Sarcoma Research GroupInstitut d'Investigació Biomèdica de Bellvitge (IDIBELL), Oncobell, L'Hospitalet de LlobregatBarcelonaSpain
| | - Silvia Mateo‐Lozano
- Developmental Tumor Biology LaboratoryInstitut de Recerca Sant Joan de Déu, Hospital Sant Joan de DéuBarcelonaSpain
- Pediatric Cancer Center BarcelonaHospital Sant Joan de DéuBarcelonaSpain
| | - Florencia Cidre‐Aranaz
- Division of Translational Paediatric Sarcoma ResearchGerman Cancer Research Center (DKFZ), German Cancer Consortium (DKTK)HeidelbergGermany
- Hopp‐Children's Cancer Center (KiTZ)HeidelbergGermany
- National Center for Tumor Diseases (NCT)NCT Heidelberg, a partnership between DKFZ and Heidelberg University HospitalGermany
| | - Thomas G.P. Grünewald
- Division of Translational Paediatric Sarcoma ResearchGerman Cancer Research Center (DKFZ), German Cancer Consortium (DKTK)HeidelbergGermany
- Hopp‐Children's Cancer Center (KiTZ)HeidelbergGermany
- National Center for Tumor Diseases (NCT)NCT Heidelberg, a partnership between DKFZ and Heidelberg University HospitalGermany
- Institute of PathologyHeidelberg University HospitalHeidelbergGermany
| | - Ángel Díaz‐Lagares
- Epigenomics Unit, Cancer Epigenomics, Translational Medical Oncology (ONCOMET), Health Research Institute of Santiago (IDIS)University Clinical Hospital of Santiago (CHUS/SERGAS)Santiago de CompostelaSpain
- Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry SchoolUniversidade de Santiago de Compostela (USC)Spain
- Universidad de Santiago de Compostela (USC)Spain
- Department of Clinical AnalysisUniversity Hospital Complex of Santiago de Compostela (CHUS)Spain
- CIBERONCCarlos III Institute of Health (ISCIII)MadridSpain
| | - Roser Lopez‐Alemany
- Sarcoma Research GroupInstitut d'Investigació Biomèdica de Bellvitge (IDIBELL), Oncobell, L'Hospitalet de LlobregatBarcelonaSpain
| | - Òscar M. Tirado
- Sarcoma Research GroupInstitut d'Investigació Biomèdica de Bellvitge (IDIBELL), Oncobell, L'Hospitalet de LlobregatBarcelonaSpain
- CIBERONCCarlos III Institute of Health (ISCIII)MadridSpain
- Institut Català d'Oncologia (ICO)L'Hospitalet de LlobregatBarcelonaSpain
| |
Collapse
|
49
|
Krause MJ, Sinkala M, Ramesar R. Distinct dysregulated pathways in sporadic and Lynch syndrome-associated colorectal cancer offer insights for targeted treatment. FEBS Lett 2025; 599:1006-1028. [PMID: 39973357 PMCID: PMC11995676 DOI: 10.1002/1873-3468.70010] [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: 11/13/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/21/2025]
Abstract
Lynch syndrome (LS) is a hereditary disorder that increases the risk of colorectal cancer (CRC) due to constitutional pathogenic variants in mismatch repair (MMR) genes. When coupled with somatic mutations in the same gene, MMR deficiency occurs. However, the mechanisms driving cancer development remain unclear. This study aimed to identify distinct molecular drivers in LS-associated and sporadic CRC. We found that PI3K-Akt signalling is dysregulated in LS-associated CRC, while Wnt signalling predominates in sporadic CRC. Moreover, our findings highlight the therapeutic potential of PI3K-Akt pathway inhibitors, such as taselisib, for LS-associated CRC patients with high pathway dependency. Similarly, Wnt signalling pathway inhibitors, such as XAV939, offer a promising therapeutic approach for sporadic CRC. These findings underscore the importance of understanding the biological basis of disease for developing targeted therapies tailored to CRC subtype-specific oncogenic pathways.
Collapse
Affiliation(s)
- May J. Krause
- UCT MRC Genomic and Precision Medicine Research Unit, Division of Human Genetics, Department of Pathology, Institute of Infectious Diseases and Molecular MedicineFaculty of Health Sciences, University of Cape TownSouth Africa
| | - Musalula Sinkala
- Computational Biology Division, Department of Integrative Biomedical Sciences, School of Health SciencesUniversity of Cape TownSouth Africa
| | - Raj Ramesar
- UCT MRC Genomic and Precision Medicine Research Unit, Division of Human Genetics, Department of Pathology, Institute of Infectious Diseases and Molecular MedicineFaculty of Health Sciences, University of Cape TownSouth Africa
| |
Collapse
|
50
|
Neves FR, Martins AL, Oliveira RC, Martins R. Characterization of the Pancreatic Neuroendocrine Neoplasm Immune Microenvironment. Cancer Med 2025; 14:e70798. [PMID: 40145271 PMCID: PMC11947738 DOI: 10.1002/cam4.70798] [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: 12/17/2024] [Revised: 03/01/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
INTRODUCTION A tumor is composed of more than tumoral cells. In recent years, there has been an increase in interest and knowledge of the tumor microenvironment (TME). METHODS The TME is an integral part of the tumor, composed of several cells: immune, stromal, and endothelial, among others, thus offering a wide range of tumor interactions and multiple possibilities for targeted therapies and environment modulation. While the TME in pancreatic ductal adenocarcinoma is widely studied, it is not very true for the TME of pancreatic neuroendocrine neoplasms (PNENs). DISCUSSION AND CONCLUSION The incidence of PNENs is increasing and, therefore, it is important to comprehend their biology for the evolution of efficient therapies since many of the PNENs develop metastasis, including the G1 PNENs. This paper focuses on a review of the role of the TME in PNENs.
Collapse
Affiliation(s)
| | | | | | - Rui Martins
- Instituto Português de Oncologia de CoimbraCoimbraPortugal
- Faculdade de Medicina da Universidade de CoimbraCoimbraPortugal
| |
Collapse
|