1
|
Giovarelli M, Mocciaro E, Carnovale C, Cervia D, Perrotta C, Clementi E. Immunosenescence in skeletal muscle: The role-play in cancer cachexia chessboard. Semin Cancer Biol 2025; 111:48-59. [PMID: 40020976 DOI: 10.1016/j.semcancer.2025.02.012] [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: 12/23/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/03/2025]
Abstract
With the increase in life expectancy, age-related conditions and diseases have become a widespread and relevant social burden. Among these, immunosenescence and cancer cachexia play a significant often intertwined role. Immunosenescence is the progressive aging decline of both the innate and adaptive immune systems leading to increased infection susceptibility, poor vaccination efficacy, autoimmune disease, and malignancies. Cancer cachexia affects elderly patients with cancer causing severe weight loss, muscle wasting, inflammation, and reduced response to therapies. Whereas the connections between immunosenescence and cancer cachexia have been raising attention, the molecular mechanisms still need to be completely elucidated. This review aims at providing the current knowledge about the interplay between immunosenescence, skeletal muscle, and cancer cachexia, analyzing the molecular pathways known so far to be involved. Finally, we highlight potential therapeutic strategies suited for elderly population aimed to block immunosenescence and to preserve muscle mass in cachexia, also presenting the analysis of the current state-of-the-art of related clinical trials.
Collapse
Affiliation(s)
- Matteo Giovarelli
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy.
| | - Emanuele Mocciaro
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy
| | - Carla Carnovale
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, Viterbo 01100, Italy
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy.
| |
Collapse
|
2
|
Wu Y, Jia N, Sun J, Liao W, Xu J, Chen W, Zhao C. The roles of algal polysaccharides in modulating tumor immune microenvironment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156610. [PMID: 40085993 DOI: 10.1016/j.phymed.2025.156610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/26/2025] [Accepted: 03/02/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Polysaccharides from algae provide a range of biology and health benefits. Lately, there has been a significant interest in how algal polysaccharides affect the immune microenvironment around tumors. PURPOSE To elucidate the subtle interactions between algal polysaccharides and the tumor immune microenvironment to further understand the medicinal potential of algal polysaccharides. STUDY DESIGN To give a summary of the sources, bioactivities and characteristics of the tumor immune microenvironment of algal polysaccharides, and to analyze alteration of the immunological milieu surrounding tumors by algal polysaccharides and their potential as immunomodulators of chemotherapeutic agents. METHODS Search popular academic search engines using selected keywords for articles ending before September 2024 using selected keywords Google Scholar, PubMed, ScienceDirect, Scopus, Web of Science, Springer, and official websites. RESULTS Algal polysaccharides can fight tumors by changing how immune cells work and affecting inflammation in different ways. Moreover, algal polysaccharides have shown promise in mitigating the adverse effects associated with conventional cancer treatments, such as chemotherapy. Algal polysaccharides, through their immunomodulatory effects, can alleviate some of these side effects, leading to an enhanced overall treatment outcome. CONCLUSION As research continues to uncover the underlying mechanisms of their antitumor effects, algal polysaccharides are poised to become a vital component in the development of novel cancer treatments, providing new hope for patients and advancing the field of oncology.
Collapse
Affiliation(s)
- Yinfeng Wu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China
| | - Nan Jia
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China
| | - Jingyu Sun
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China
| | - Wei Liao
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China
| | - Jingxiang Xu
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, PR China
| | - Weichao Chen
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China
| | - Chao Zhao
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China.
| |
Collapse
|
3
|
Kao C, Charmsaz S, Tsai HL, Aziz K, Shu DH, Munjal K, Griffin E, Leatherman JM, Lipson EJ, Ged Y, Hoffman-Censits J, Li HL, Hallab E, Brancati M, Nakazawa M, Alden S, Thoburn C, Gross NE, Hernandez AG, Coyne EM, Kartalia E, Baretti M, Jaffee EM, Bansal S, Tang L, Chandler GS, Mohindra R, Ho WJ, Yarchoan M, Zabransky DJ. Age-related divergence of circulating immune responses in patients with solid tumors treated with immune checkpoint inhibitors. Nat Commun 2025; 16:3531. [PMID: 40258833 PMCID: PMC12012091 DOI: 10.1038/s41467-025-58512-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 03/19/2025] [Indexed: 04/23/2025] Open
Abstract
Most new cancer diagnoses occur in patients over the age of 65. The composition and function of the immune system changes with age, but how the aged immune system affects responses to immune checkpoint inhibitor (ICI) cancer therapies remains incompletely understood. Here, using multiplex cytokine assay and high-parameter mass cytometry, we analyze prospectively collected blood samples from 104 cancer patients receiving ICIs. We find aged patients ( ≥ 65-years-old; n = 54) derive similar clinical outcomes as younger patients (n = 50). However, aged, compared to young, patients have divergent immune phenotypes at baseline that persist during ICI therapy, including diminished cytokine responses, reduced pools of naïve T cells with increased relative expression of immune checkpoint molecules, and more robust effector T cell expansion in responders compared to non-responders. Our study provides insights into age-stratified mechanisms of ICI effects while also implying the utility of age-tailored immunotherapeutic approaches.
Collapse
Affiliation(s)
- Chester Kao
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Soren Charmsaz
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hua-Ling Tsai
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Khaled Aziz
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel H Shu
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kabeer Munjal
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ervin Griffin
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - James M Leatherman
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Evan J Lipson
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yasser Ged
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Howard L Li
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elsa Hallab
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Madelena Brancati
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mari Nakazawa
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephanie Alden
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher Thoburn
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicole E Gross
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexei G Hernandez
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Erin M Coyne
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emma Kartalia
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marina Baretti
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth M Jaffee
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University, Baltimore, MD, USA
| | | | - Laura Tang
- Genentech Inc, South San Francisco, California, USA
| | | | | | - Won Jin Ho
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Convergence Institute, Johns Hopkins University, Baltimore, MD, USA.
| | - Mark Yarchoan
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Convergence Institute, Johns Hopkins University, Baltimore, MD, USA.
| | - Daniel J Zabransky
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Convergence Institute, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
4
|
Afzal A, Abbasi MH, Ahmad S, Sheikh N, Khawar MB. Current Trends in Messenger RNA Technology for Cancer Therapeutics. Biomater Res 2025; 29:0178. [PMID: 40207255 PMCID: PMC11978394 DOI: 10.34133/bmr.0178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 04/11/2025] Open
Abstract
Messenger RNA (mRNA)-based therapy has revolutionized cancer research by enabling versatile delivery systems for therapeutic applications. The future of mRNA-based cancer therapies shows promise amidst challenges such as delivery efficiency, immunogenicity, and tumor heterogeneity. Recent progress has adapted various strategies such as design flexibility, scalable production, and targeted delivery capabilities to enhance the potential in personalized cancer therapy. Further research to optimize delivery for enhanced outcomes and efficacy in solid tumors is warranted. Therefore, we aim to explore the current landscape and future prospects of mRNA technology across various therapeutic platforms.
Collapse
Affiliation(s)
- Ali Afzal
- Applied Molecular Biology and Biomedicine Lab, Department of Zoology,
University of Narowal, Narowal, Pakistan
| | | | - Shaaf Ahmad
- King Edward Medical University/Mayo Hospital, Lahore, Punjab 54000, Pakistan
| | - Nadeem Sheikh
- Cell & Molecular Biology Lab, Institute of Zoology,
University of the Punjab, Lahore, Pakistan
| | - Muhammad Babar Khawar
- Applied Molecular Biology and Biomedicine Lab, Department of Zoology,
University of Narowal, Narowal, Pakistan
| |
Collapse
|
5
|
Nagy G, Gunkl-Tóth L, Dorgó AM, McInnes IB. The concept of difficult-to-treat disease in rheumatology: where next? THE LANCET. RHEUMATOLOGY 2025; 7:e274-e289. [PMID: 39848270 DOI: 10.1016/s2665-9913(24)00340-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 01/25/2025]
Abstract
New pathogenesis-based therapeutics and evidence-based consensus treatment recommendations, often with predefined treatment goals, have remarkably improved outcomes across many chronic diseases. However, a clinically significant subgroup of patients responds poorly to interventions and show a progressive decline in the disease trajectory, which poses an increasing health-care challenge. Difficult-to-treat approaches exist in several areas of medicine and the need for similar definitions has recently also emerged in rheumatology. The term difficult-to-treat refers not only to patients with pathology-driven, treatment-refractory disease, but also implicates multiple other factors that can contribute to patients being in this state, including having few treatment options, misdiagnosis, and coincident psychosocial factors. Therefore, the difficult-to-treat state requires a comprehensive, holistic, multidisciplinary approach that considers the specific characteristics of each disease and the personalised needs of the patient. In this Personal View, we provide an overview of the different aspects of the concept of difficult-to-treat disease, highlight its advantages, and propose the importance of incorporating this concept more widely in the design of rheumatological treatment strategies.
Collapse
Affiliation(s)
- György Nagy
- National Institute of Locomotor Diseases and Disabilities, Budapest, Hungary; Department of Rheumatology and Clinical Immunology, Semmelweis University, Budapest, Hungary; Department of Internal Medicine and Oncology and Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary; Heart and Vascular Center, Semmelweis University, Budapest, Hungary.
| | - Lilla Gunkl-Tóth
- Department of Rheumatology and Clinical Immunology, Semmelweis University, Budapest, Hungary; Department of Pharmacology and Pharmacotherapy, University of Pécs, Pécs, Hungary; Hungarian Research Network Chronic Pain Research Group, Pécs, Hungary
| | - András M Dorgó
- Department of Rheumatology and Clinical Immunology, Semmelweis University, Budapest, Hungary
| | - Iain B McInnes
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
6
|
Nishida A, Andoh A. The Role of Inflammation in Cancer: Mechanisms of Tumor Initiation, Progression, and Metastasis. Cells 2025; 14:488. [PMID: 40214442 PMCID: PMC11987742 DOI: 10.3390/cells14070488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/14/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
Inflammation is an essential component of the immune response that protects the host against pathogens and facilitates tissue repair. Chronic inflammation is a critical factor in cancer development and progression. It affects every stage of tumor development, from initiation and promotion to invasion and metastasis. Tumors often create an inflammatory microenvironment that induces angiogenesis, immune suppression, and malignant growth. Immune cells within the tumor microenvironment interact actively with cancer cells, which drives progression through complex molecular mechanisms. Chronic inflammation is triggered by factors such as infections, obesity, and environmental toxins and is strongly linked to increased cancer risk. However, acute inflammatory responses can sometimes boost antitumor immunity; thus, inflammation presents both challenges and opportunities for therapeutic intervention. This review examines how inflammation contributes to tumor biology, emphasizing its dual role as a critical factor in tumorigenesis and as a potential therapeutic target.
Collapse
Affiliation(s)
- Atsushi Nishida
- Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu 520-2192, Shiga, Japan;
| | | |
Collapse
|
7
|
Semeradtova A, Liegertova M, Herma R, Capkova M, Brignole C, Del Zotto G. Extracellular vesicles in cancer´s communication: messages we can read and how to answer. Mol Cancer 2025; 24:86. [PMID: 40108630 PMCID: PMC11921637 DOI: 10.1186/s12943-025-02282-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Extracellular vesicles (EVs) are emerging as critical mediators of intercellular communication in the tumor microenvironment (TME), profoundly influencing cancer progression. These nano-sized vesicles, released by both tumor and stromal cells, carry a diverse cargo of proteins, nucleic acids, and lipids, reflecting the dynamic cellular landscape and mediating intricate interactions between cells. This review provides a comprehensive overview of the biogenesis, composition, and functional roles of EVs in cancer, highlighting their significance in both basic research and clinical applications. We discuss how cancer cells manipulate EV biogenesis pathways to produce vesicles enriched with pro-tumorigenic molecules, explore the specific contributions of EVs to key hallmarks of cancer, such as angiogenesis, metastasis, and immune evasion, emphasizing their role in shaping TME and driving therapeutic resistance. Concurrently, we submit recent knowledge on how the cargo of EVs can serve as a valuable source of biomarkers for minimally invasive liquid biopsies, and its therapeutic potential, particularly as targeted drug delivery vehicles and immunomodulatory agents, showcasing their promise for enhancing the efficacy and safety of cancer treatments. By deciphering the intricate messages carried by EVs, we can gain a deeper understanding of cancer biology and develop more effective strategies for early detection, targeted therapy, and immunotherapy, paving the way for a new era of personalized and precise cancer medicine with the potential to significantly improve patient outcomes.
Collapse
Affiliation(s)
- Alena Semeradtova
- Institute of Photonics and Electronics of the CAS, Chaberská 1014/57, Prague, 182 51, Czech Republic.
| | - Michaela Liegertova
- Centre for Nanomaterials and Biotechnology, Faculty of Science, Jan Evangelista Purkyně University in Ústí Nad Labem, Pasteurova 3632/15, Ústí Nad Labem, 40096, Czech Republic
| | - Regina Herma
- Centre for Nanomaterials and Biotechnology, Faculty of Science, Jan Evangelista Purkyně University in Ústí Nad Labem, Pasteurova 3632/15, Ústí Nad Labem, 40096, Czech Republic
| | - Magdalena Capkova
- Institute of Photonics and Electronics of the CAS, Chaberská 1014/57, Prague, 182 51, Czech Republic
| | - Chiara Brignole
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147, Genoa, Italy.
| | - Genny Del Zotto
- Core Facilities, Department of Research and Diagnostics, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy.
| |
Collapse
|
8
|
Vogrig A, Dentoni M, Florean I, Cellante G, Domenis R, Iacono D, Pelizzari G, Rossi S, Damato V, Fabris M, Valente M. Prediction, prevention, and precision treatment of immune checkpoint inhibitor neurological toxicity using autoantibodies, cytokines, and microbiota. Front Immunol 2025; 16:1548897. [PMID: 40181971 PMCID: PMC11966491 DOI: 10.3389/fimmu.2025.1548897] [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: 12/20/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Cancer immunotherapy with immune checkpoint inhibitors (ICIs) has revolutionized oncology, significantly improving survival across multiple cancer types. ICIs, such as anti-PD-1 (e.g. nivolumab, pembrolizumab), anti-PD-L1 (e.g. atezolizumab, avelumab), and anti-CTLA-4 (e.g. ipilimumab), enhance T cell-mediated anti-tumor responses but can also trigger immune-related adverse events (irAEs). Neurological irAEs (n-irAEs), affecting 1-3% of patients, predominantly involve the peripheral nervous system; less commonly, n-irAEs can present as central nervous system disorders. Although irAEs suggest a possible correlation with treatment efficacy, their mechanisms remain unclear, with hypotheses ranging from antigen mimicry to cytokine dysregulation and microbiome alterations. Identifying patients at risk for n-irAEs and predicting their outcome through biomarkers would be highly desirable. For example, patients with high-risk onconeural antibodies (such as anti-Hu or Ma2), and elevated neurofilament light chain (NfL) levels often respond poorly to irAE treatment. However, interpreting neuronal antibody tests in the diagnosis of n-irAEs requires caution: positive results must align with the clinical context, as some cancer patients (e.g., SCLC) may have asymptomatic low antibody levels, and false positive results are common without tissue-based confirmation. Also, the use of biomarkers (e.g. IL-6) may lead to more targeted treatments of irAEs, minimizing adverse effects without compromising the anti-tumor efficacy of ICIs. This review provides a comprehensive overview of the latest findings on n-irAEs associated with ICIs, with a focus on their prediction, prevention, as well as precision treatment using autoantibodies, cytokines, and microbiota. The most interesting data concern neuronal antibodies, which we explore in their pathogenic roles and as biomarkers of neurotoxicity. Most of the available data on cytokines, both regarding their role as diagnostic and prognostic biomarkers and their role in supporting therapeutic decisions for toxicities, refer to non-neurological toxicities. However, in our review, we mention the potential role of CXCL10 and CXCL13 as biomarkers of n-irAEs and describe the current evidence, as well as the need for further studies, on the use of cytokines in guiding selection of second-line therapies for n-irAEs. Finally, no specific microbiome-related microbial signature has been proven to be linked to n-irAEs specifically, leading to the need of more future research on the topic.
Collapse
Affiliation(s)
- Alberto Vogrig
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Clinical Neurology, Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Marta Dentoni
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Clinical Neurology, Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Irene Florean
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Clinical Neurology, Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Giulia Cellante
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Clinical Neurology, Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Rossana Domenis
- Institute of Clinical Pathology, Department of Laboratory Medicine, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Donatella Iacono
- Department of Oncology, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Giacomo Pelizzari
- Department of Oncology, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Simone Rossi
- IRCCS - Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Valentina Damato
- Department of Neurosciences, Drugs and Child Health, University of Florence, Firenze, Italy
| | - Martina Fabris
- Institute of Clinical Pathology, Department of Laboratory Medicine, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Mariarosaria Valente
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Clinical Neurology, Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| |
Collapse
|
9
|
Silinskaite U, Valciukiene J, Jakubauskas M, Poskus T. The Immune Environment in Colorectal Adenoma: A Systematic Review. Biomedicines 2025; 13:699. [PMID: 40149674 PMCID: PMC11940254 DOI: 10.3390/biomedicines13030699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Research on colorectal adenoma is significantly less comprehensive compared to studies on colorectal carcinoma. Although colorectal adenoma is a precursor of the majority of sporadic colorectal cancers, not all adenomas develop into carcinomas. The complex interaction of immune responses in the premalignant tumor microenvironment might be a factor for that. Methods: In this systematic review, we aim to provide a thorough analysis of the current research examining the immune infiltration patterns in sporadic colorectal adenoma tissues in the context of immune cell-based, cytokine-based, and other immunological factor-related changes along the conventional adenoma-carcinoma sequence. The articles included in the review extend up to December 2024 in PubMed and Web of Science databases. Results: Most included studies have shown significant differences in immune cell counts, densities, and cytokine expression levels associated with premalignant colorectal lesions (and/or colorectal cancer). No consensus on the immune-related tendencies concerning CD4+T cells and CD8+T cells was reached. Decreasing expression of mDCs and plasma and naïve B cells were detected along the ACS. The increased density of tissue eosinophils in the adenoma tissue dramatically diminishes after the transition to carcinoma. As the adenoma progresses, the increasing expression of IL-1α, IL-4, IL-6, IL-8, IL-10, IL-17A, IL-21, IL-23, IL-33, and TGF-β and decreasing levels of IL-12A, IL-18, IFN-γ, and TNFα cytokines in the invasive carcinoma stage is being detected. The over-expression of COX-2, PD-1/PD-L1, CTLA-4, and ICOS/ICOSLG in the colorectal adenomatous and cancerous tissues was also observed. Conclusions: Further studies are needed for a better understanding of the whole picture of colorectal adenoma-associated immunity and its impact on precancerous lesion's potential to progress.
Collapse
|
10
|
Tripathi S, Sharma Y, Kumar D. Unveiling the link between chronic inflammation and cancer. Metabol Open 2025; 25:100347. [PMID: 39876904 PMCID: PMC11772974 DOI: 10.1016/j.metop.2025.100347] [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: 11/10/2024] [Revised: 01/05/2025] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
The highly nuanced transition from an inflammatory process to tumorigenesis is of great scientific interest. While it is well known that environmental stimuli can cause inflammation, less is known about the oncogenic modifications that chronic inflammation in the tissue microenvironment can bring about, as well as how these modifications can set off pro-tumorigenic processes. It is clear that no matter where the environmental factors come from, maintaining an inflammatory microenvironment encourages carcinogenesis. In addition to encouraging angiogenesis and metastatic processes, sustaining the survival and proliferation of malignant transformed cells, and possibly altering the efficacy of therapeutic agents, inflammation can negatively regulate the antitumoral adaptive and innate immune responses. Because chronic inflammation has multiple pathways involved in tumorigenesis and metastasis, it has gained recognition as a marker of cancer and a desirable target for cancer therapy. Recent advances in our knowledge of the molecular mechanisms that drive cancer's progression demonstrate that inflammation promotes tumorigenesis and metastasis while suppressing anti-tumor immunity. In many solid tumor types, including breast, lung, and liver cancer, inflammation stimulates the activation of oncogenes and impairs the body's defenses against the tumor. Additionally, it alters the microenvironment of the tumor. As a tactical approach to cancer treatment, these findings have underscored the importance of targeting inflammatory pathways. This review highlights the role of inflammation in cancer development and metastasis, focusing on its impact on tumor progression, immune suppression, and therapy resistance. It examines current anti-inflammatory strategies, including NSAIDs, cytokine modulators, and STAT3 inhibitors, while addressing their potential and limitations. The review emphasizes the need for further research to unravel the complex mechanisms linking inflammation to cancer progression and identify molecular targets for specific cancer subtypes.
Collapse
Affiliation(s)
- Siddhant Tripathi
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Yashika Sharma
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Dileep Kumar
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| |
Collapse
|
11
|
Baharom F, Hermans D, Delamarre L, Seder RA. Vax-Innate: improving therapeutic cancer vaccines by modulating T cells and the tumour microenvironment. Nat Rev Immunol 2025; 25:195-211. [PMID: 39433884 DOI: 10.1038/s41577-024-01091-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2024] [Indexed: 10/23/2024]
Abstract
T cells have a critical role in mediating antitumour immunity. The success of immune checkpoint inhibitors (ICIs) for cancer treatment highlights how enhancing endogenous T cell responses can mediate tumour regression. However, mortality remains high for many cancers, especially in the metastatic setting. Based on advances in the genetic characterization of tumours and identification of tumour-specific antigens, individualized therapeutic cancer vaccines targeting mutated tumour antigens (neoantigens) are being developed to generate tumour-specific T cells for improved therapeutic responses. Early clinical trials using individualized neoantigen vaccines for patients with advanced disease had limited clinical efficacy despite demonstrated induction of T cell responses. Therefore, enhancing T cell activity by improving the magnitude, quality and breadth of T cell responses following vaccination is one current goal for improving outcome against metastatic tumours. Another major consideration is how T cells can be further optimized to function within the tumour microenvironment (TME). In this Perspective, we focus on neoantigen vaccines and propose a new approach, termed Vax-Innate, in which vaccination through intravenous delivery or in combination with tumour-targeting immune modulators may improve antitumour efficacy by simultaneously increasing the magnitude, quality and breadth of T cells while transforming the TME into a largely immunostimulatory environment for T cells.
Collapse
Affiliation(s)
| | - Dalton Hermans
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA
| | | | - Robert A Seder
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
12
|
Wang Z, Dai W, Zhang Z, Wang H. Aptamer-Based Graphene Field-Effect Transistor Biosensor for Cytokine Detection in Undiluted Physiological Media for Cervical Carcinoma Diagnosis. BIOSENSORS 2025; 15:138. [PMID: 40136934 PMCID: PMC11939848 DOI: 10.3390/bios15030138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025]
Abstract
Personalized monitoring of disease biomarkers is of great interest in women's health. However, existing approaches typically involve invasive inspection or bulky equipment, making them challenging to implement at home. Hence, we present a general strategy for label-free and specific detection of disease biomarkers in physiological media using an aptamer-based biosensor. The biosensor is a graphene field-effect transistor that involves immobilizing the aptamer and a biomolecule-permeable polyethylene glycol (PEG) layer on the graphene surface. The aptamer is capable of specifically binding with the target biomarker, thus inducing a change in the sensing responses. The PEG layer can effectively reduce the nonspecific adsorption of nontarget molecules in the solution, and increase the effective Debye screening length in the region directly adjacent to the graphene. In this work, studies of a biosensor with modification of the aptamer and PEG show that cervical carcinoma biomarkers such as tumor necrosis factor-α and interleukin 6 can be sensitively and specifically detected in undiluted physiological media, with detection limits as low as 0.13 pM for TNF-a and 0.20 pM for IL-6. This work presents a significant method for the general application of the biosensor for disease diagnosis in women's health.
Collapse
Affiliation(s)
- Ziran Wang
- Key Laboratory of High-Efficiency and Clean Mechanical Manufacture of MOE, School of Mechanical Engineering, Shandong University, Jinan 250100, China
| | - Wenting Dai
- Department of Mechanical Engineering, Columbia University, New York, NY 10027, USA
| | - Zaiyu Zhang
- Key Laboratory of High-Efficiency and Clean Mechanical Manufacture of MOE, School of Mechanical Engineering, Shandong University, Jinan 250100, China
| | - Haipeng Wang
- Key Laboratory of High-Efficiency and Clean Mechanical Manufacture of MOE, School of Mechanical Engineering, Shandong University, Jinan 250100, China
| |
Collapse
|
13
|
Ciurescu S, Tomescu L, Șerban D, Nicolae N, Nan G, Buciu V, Ilaș DG, Cîtu C, Vernic C, Sas I. The Prognostic Value of Systemic Inflammation Index in Breast Cancer: A Retrospective Study in Western Romania. J Clin Med 2025; 14:1081. [PMID: 40004612 PMCID: PMC11856417 DOI: 10.3390/jcm14041081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 01/30/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Breast cancer remains a leading cause of cancer-related morbidity worldwide, and refining prognostic tools is essential for individualized patient management. Recent evidence suggests that the systemic immune-inflammation index (SII), derived from routine blood tests, may offer valuable prognostic insights. This study aimed to evaluate whether SII can reliably predict clinical outcomes for patients undergoing curative resection. Methods: We retrospectively analyzed patients with histologically confirmed breast cancer who underwent surgical intervention at a single tertiary center. Preoperative complete blood counts were used to calculate SII. Using receiver operating characteristic (ROC) curve analysis, we identified an optimal SII cutoff. Using statistical tests, including t-tests and ANOVA, we examined differences in clinicopathological factors between low- and high-SII groups. Using univariate and multivariate analyses, we explored associations between SII and variables such as tumor stage and hormone receptor status. Results: Patients with elevated SII levels showed significant associations with more advanced tumor stage and systemic inflammatory profiles. The identified SII cutoff separated patients into distinct risk groups, and high SII values correlated with poorer prognostic features. Multivariate models indicated that SII provided additional predictive value beyond standard markers. Conclusions: Our findings suggest that SII may provide prognostic insights into breast cancer, particularly in stratifying patients based on inflammatory profiles. However, the current study does not support the use of SII as a clinical tool for tailoring treatment strategies. Further preclinical and randomized controlled trials are required to determine its predictive utility and to assess its potential integration into personalized management approaches.
Collapse
Affiliation(s)
- Sebastian Ciurescu
- Doctoral School in Medicine, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania; (S.C.); (G.N.); (V.B.)
- Department of Obstetrics and Gynecology, Victor Babeş University of Medicine and Pharmacy, 300041 Timișoara, Romania; (L.T.); (D.Ș.); (N.N.); (C.C.); (I.S.)
| | - Larisa Tomescu
- Department of Obstetrics and Gynecology, Victor Babeş University of Medicine and Pharmacy, 300041 Timișoara, Romania; (L.T.); (D.Ș.); (N.N.); (C.C.); (I.S.)
| | - Denis Șerban
- Department of Obstetrics and Gynecology, Victor Babeş University of Medicine and Pharmacy, 300041 Timișoara, Romania; (L.T.); (D.Ș.); (N.N.); (C.C.); (I.S.)
| | - Nicoleta Nicolae
- Department of Obstetrics and Gynecology, Victor Babeş University of Medicine and Pharmacy, 300041 Timișoara, Romania; (L.T.); (D.Ș.); (N.N.); (C.C.); (I.S.)
| | - Georgiana Nan
- Doctoral School in Medicine, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania; (S.C.); (G.N.); (V.B.)
- Department of Obstetrics and Gynecology, Victor Babeş University of Medicine and Pharmacy, 300041 Timișoara, Romania; (L.T.); (D.Ș.); (N.N.); (C.C.); (I.S.)
| | - Victor Buciu
- Doctoral School in Medicine, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania; (S.C.); (G.N.); (V.B.)
- Department of Obstetrics and Gynecology, Victor Babeş University of Medicine and Pharmacy, 300041 Timișoara, Romania; (L.T.); (D.Ș.); (N.N.); (C.C.); (I.S.)
| | - Diana-Gabriela Ilaș
- Department of Medical Semiology, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Cosmin Cîtu
- Department of Obstetrics and Gynecology, Victor Babeş University of Medicine and Pharmacy, 300041 Timișoara, Romania; (L.T.); (D.Ș.); (N.N.); (C.C.); (I.S.)
| | - Corina Vernic
- Doctoral School in Medicine, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania; (S.C.); (G.N.); (V.B.)
- Department of Medical Informatics and Biostatistics, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Ioan Sas
- Department of Obstetrics and Gynecology, Victor Babeş University of Medicine and Pharmacy, 300041 Timișoara, Romania; (L.T.); (D.Ș.); (N.N.); (C.C.); (I.S.)
| |
Collapse
|
14
|
Refaat S, Al-Rashidi HE, El Azeem RAA, Nouh WE, Hamed S, Attia ZR. The functional TNF-α -308G > a single-nucleotide polymorphism (rs1800629): association with the predictive indices of breast cancer carcinogenesis. Breast Cancer Res Treat 2025; 210:57-70. [PMID: 39570546 PMCID: PMC11787156 DOI: 10.1007/s10549-024-07536-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Compared with all other cancer types, Breast cancer (BC) among women has now exceeded them all as the primary reason for cancer worldwide. The BC represents 11.7% of all cancer cases and accounts for a predestined 2.3 million new cases. It is the fourth primary reason for cancer-associated deaths in women. With a staggering 200-400% increase in the relative incidence of BC in Egypt, there is an urgent need for new diagnostic or predictive markers. PURPOSE The current investigation aims to explore the connection of the functional TNF-α-308G > A (rs1800629) single-nucleotide polymorphism (SNP) with different breast cancer predictive indices. METHODS The ARMS-PCR method was used for genotyping TNF-α-308G > A SNP. Three groups were recruited for the study: 79 patients with benign breast inflammation (BBI); 163 with breast cancer (BC) and 144 controls (C). RESULTS The TNF-α-308G > A SNP was distributed among different groups in a unique pattern; in the control group 63.9% of cases were in the GG, 34% were in the GA, and 2.1% were in the AA. The BC group had 14% GG, 79% GA, and 7% AA, while the BBI group had 24% GG, 76% GA, and 0% AA. The AA genotype and A allele represented a strong significant correlation with risk factors in the BC group (ORAA: 14.67 [95% CI = 3.78-56.91] and ORA: 0.27 [95% CI = 0.19-0.39], respectively; P < 0.0001) in contrast to the control group. However, in the BBI group, a strong significant correlation was noted with the GA genotype (ORGA: 5.93 [95% CI = 3.18-11.04] P < 0.0001). In the BC group, the AA genotype shows a significant increase in Nottingham Prognostic Index (NPI) in positive ER and PR in contrast to the relevant negative ones (P = 0.02 and 0.002, respectively). However, the GA genotype significantly increased NPI in positive Her2 and metastatic patients (P = 0.03 and 0.01, respectively). CONCLUSION This research is the first to correlate TNF-α-308G > A (rs1800629) SNP in Egyptian BC patients. The A allele, GA & AA genotypes, and the Overdominant model of the TNF-α-308G > A gene variants were recorded as prognostic risk factors for BC carcinogenesis.
Collapse
Affiliation(s)
- Sherif Refaat
- Department of Medical Oncology, Oncology Center, Mansoura University, Mansoura, Egypt
| | - Hanan E Al-Rashidi
- Medical Laboratory Technology Department, College of Applied Medical Science, Taibah University, Madinah, Saudi Arabia
| | - Rania A Abd El Azeem
- Mansoura University Children's Hospital, Mansoura University, Mansoura, Egypt
- Department of Clinical Laboratory Sciences, College of Medical Applied Sciences, University of Hafr Al Batin, Hafr Al Batin, Saudi Arabia
| | - Walaa E Nouh
- Mansoura University Children's Hospital, Mansoura University, Mansoura, Egypt
| | - Sahar Hamed
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt.
| | - Zeinab R Attia
- Mansoura University Children's Hospital, Mansoura University, Mansoura, Egypt
| |
Collapse
|
15
|
Qin Y, Dong X, Li B. Salivary miRNAs and cytokines associated with diagnosis and prognosis of oral squamous cell carcinoma. Front Cell Dev Biol 2025; 13:1531016. [PMID: 39911325 PMCID: PMC11794800 DOI: 10.3389/fcell.2025.1531016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 01/07/2025] [Indexed: 02/07/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common malignant tumour in the oral and maxillofacial region. Early diagnosis can significantly improve the 5-year survival rate of patients with OSCC. Therefore, it is extremely important to differentiate OSCC patients early, easily and quickly. Human saliva contains a variety of components that can be used as biomarkers for the diagnosis and prognosis of OSCC. Studies have shown that salivary microRNAs (miRNAs) and cytokines are closely associated with the progression of OSCC. The aim of this review is to summarize the research progress of salivary biomarkers (miRNAs and cytokines) in the past 3 years, and to explore the possibility of using miRNAs and cytokines to improve the diagnosis and prognosis of OSCC.
Collapse
Affiliation(s)
| | | | - Bo Li
- Department of Oral Anatomy and Physiology, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
16
|
Iancu D, Fulga A, Vesa D, Fulga I, Tutunaru D, Zenovia A, Piraianu AI, Stamate E, Sterian C, Dimofte F, Badea MA, Tatu AL. Immunosuppression and Outcomes in Patients with Cutaneous Squamous Cell Carcinoma of the Head and Neck. Clin Pract 2025; 15:21. [PMID: 39851804 PMCID: PMC11764338 DOI: 10.3390/clinpract15010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
Cutaneous squamous scell carcinoma (cSCC) is a frequent non-melanoma skin cancer that originates from keratinocytes with increased prevalence. cSCC can be either in situ, as in Bowen's disease, or extended. Advanced age, accumulated sun exposure, light pigmentation, and prior skin cancer diagnosis are all significant risk factors for cSCC. Although most cSCCs can be treated surgically, some recur and metastasize, resulting in death. The role of immune status is not yet determined in the prognosis of these patients. Objective. Immunosuppressed patients are more likely to develop cSCC, which is often characterized by more aggressive, multifocal lesions. This study aimed to determine the risks of mortality in patients with cSCC and immunosuppression versus non immunosuppression and to compare variations in overall survival based on different clinical features. Method. We evaluated clinical cases of patients at "Sfantul Apostol Andrei" Emergency Hospital of Galati, Romania, from 1 March 2018 to 1 April 2024. Subjects in the trial had to be at least 18 years old and have a pathologically confirmed diagnosis of cutaneous head and neck squamous cell carcinoma (cHNSCC). We divided the patients into two different categories based on whether they had immunosuppression. Results. In this cohort of 68 subjects with cSCC, patients with immunosuppression had significantly lower overall survival, as well as lower three- and five-year survival rates compared with those without immunosuppression, even after adjustment for age, sex, stage, and previous surgical treatment. The median survival time for immunosuppressed individuals ranged from 11 to 21 months, varying based on their particular characteristics, and most critically, on the presence of other malignancies, while that of immunocompetent patients ranged from 18 to 51 months. In addition, immune-deficient patients with early-stage disease had a 21-month median survival rate that changed to11 months for advanced-stage cases. In a similar manner, immunocompetent patients with early-stage cancer had a significantly better median survival than those withadvancedstages,43 versus 18months. Our results indicate that immunosuppression is a distinct risk factors associated with a less favorable outcome in patients with cHNSCC.
Collapse
Affiliation(s)
- Doriana Iancu
- ENT Department, “Sfantul Andrei” Emergency Hospital of Galati, 800578 Galati, Romania
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 800010 Galati, Romania
| | - Ana Fulga
- ENT Department, “Sfantul Andrei” Emergency Hospital of Galati, 800578 Galati, Romania
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 800010 Galati, Romania
| | - Doina Vesa
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 800010 Galati, Romania
| | - Iuliu Fulga
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 800010 Galati, Romania
- Department of Forensic Medicine, “Sfantul Andrei” Emergency Clinical Hospital of Galati, 800223 Galati, Romania
| | - Dana Tutunaru
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 800010 Galati, Romania
| | - Andrei Zenovia
- Department of Otorhinolaryngology, “Cai Ferate” General Hospital, 800223 Galati, Romania
| | - Alin Ionut Piraianu
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 800010 Galati, Romania
| | - Elena Stamate
- Cardiology Department, Emergency University Hospital of Bucharest, 050098 Bucharest, Romania
| | | | - Florentin Dimofte
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 800010 Galati, Romania
| | - Mihail Alexandru Badea
- Department of Microbiology, George Emil Palade University of Medicine, Pharmacy, 540139 Targu Mures, Romania
| | - Alin Laurentiu Tatu
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 800010 Galati, Romania
- Department of Dermatology, “Sfanta Cuvioasa Parascheva” Hospital of Infectious Diseases, 800179 Galati, Romania
- Multidisciplinary Integrative Center for Dermatologic Interface Research, 800179 Galati, Romania
| |
Collapse
|
17
|
Mella C, Tsarouhas P, Brockwell M, Ball HC. The Role of Chronic Inflammation in Pediatric Cancer. Cancers (Basel) 2025; 17:154. [PMID: 39796780 PMCID: PMC11719864 DOI: 10.3390/cancers17010154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/31/2024] [Accepted: 01/01/2025] [Indexed: 01/13/2025] Open
Abstract
Inflammation plays a crucial role in wound healing and the host immune response following pathogenic invasion. However, unresolved chronic inflammation can result in tissue fibrosis and genetic alterations that contribute to the pathogenesis of human diseases such as cancer. Recent scientific advancements exploring the underlying mechanisms of malignant cellular transformations and cancer progression have exposed significant disparities between pediatric and adult-onset cancers. For instance, pediatric cancers tend to have lower mutational burdens and arise in actively developing tissues, where cell-cycle dysregulation leads to gene, chromosomal, and fusion gene development not seen in adult-onset counterparts. As such, scientific findings in adult cancers cannot be directly applied to pediatric cancers, where unique mutations and inherent etiologies remain poorly understood. Here, we review the role of chronic inflammation in processes of genetic and chromosomal instability, the tumor microenvironment, and immune response that result in pediatric tumorigenesis transformation and explore current and developing therapeutic interventions to maintain and/or restore inflammatory homeostasis.
Collapse
Affiliation(s)
- Christine Mella
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA;
| | - Panogiotis Tsarouhas
- Department of Biology, The University of Akron, 302 Buchtel Common, Akron, OH 44325, USA;
| | - Maximillian Brockwell
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA;
| | - Hope C. Ball
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA;
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA;
- Rebecca D. Considine Research Institute, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA
| |
Collapse
|
18
|
Sel FA, Oğuz FS. Cancer and Secretomes: HLA-G and Cancer Puzzle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1479:165-179. [PMID: 39841384 DOI: 10.1007/5584_2024_843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Among the mechanisms, cancer cells develop to elude immune system, immune regulation and the use of molecules that play important roles in immune escape stand out. One of these molecules, the human leukocyte antigen G (HLA-G), plays an important role in the maintenance of immune tolerance and contributes to the progression of cancer by exerting an immunosuppressive effect. By creating an immunosuppressive field in the microscopic environment of the tumor, the aberrant expression of HLA-G facilitates the evading of cancer cells from the immune system and contributes to the progression of the disease. It is important to study how HLA-Gs interact with secretome components, especially at the level of specific components, to develop treatment strategies that prevent cancer cells evading the immune system. Cancer cells may be recognized and targeted by the immune system by reducing the inhibitory effect of HLA-G on immune cells and by neutralizing tumor-promoting components of the secretome. This review focuses on the interaction of specific cancer cell secretomes and HLA-G. Here we also investigate the role of this interaction in tumor immune escape strategies.
Collapse
Affiliation(s)
- Figen Abatay Sel
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey.
| | - Fatma Savran Oğuz
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
19
|
Lopez-Pastorini A, Tatli Z, von Bargen A, Faltenberg D, Beling H, Galetin T, Koryllos A, Stoelben E. The Prognostic Value of Preoperative C-Reactive Protein Levels in Resected Early-Stage Lung Cancer. J Surg Res 2025; 305:85-92. [PMID: 39662214 DOI: 10.1016/j.jss.2024.11.003] [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: 12/23/2023] [Revised: 09/27/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024]
Abstract
INTRODUCTION C-reactive protein (CRP) is the most widely used marker of the systemic inflammatory response. An association between preoperative elevated levels and prognosis has been demonstrated for numerous tumors. The aim of this study was to investigate the association between preoperative CRP levels and survival in early-stage nonsmall cell lung cancer. METHODS Data from 915 consecutive patients who underwent complete resection for stage I and II nonsmall cell lung cancer were retrospectively analyzed. Recurrence-free survival (RFS) and overall survival (OS) according to preoperative CRP levels were evaluated by the Kaplan-Meier method. The Cox proportional hazards model and logistic regression analysis were used for multivariate analysis. RESULTS Five-year RFS and OS were 61.0% and 70.3% in the low CRP group (<4 mg/L) and 41.8% and 49.4% in the high CRP group (≥4 mg/L), respectively (P < 0.001). In univariate analysis, CRP levels were correlated with indicators of tumor burden and pulmonary comorbidity. In multivariate analysis, CRP levels were identified as an independent predictor of RFS and OS. CONCLUSIONS Elevated preoperative CRP is associated with poor prognosis in patients with early-stage lung cancer. CRP may guide risk-adapted follow-up and adjuvant therapy decisions. As CRP elevation is also associated with nontumor related conditions patients need to be screened for coexisting comorbidities.
Collapse
Affiliation(s)
- Alberto Lopez-Pastorini
- Department of Thoracic Surgery, Cologne-Merheim Hospital, Witten/Herdecke University Hospital, Cologne, Germany; Faculty of Medicine, Witten/Herdecke University, Witten, Germany.
| | - Zehra Tatli
- Faculty of Medicine, University of Cologne, Cologne, Germany
| | | | | | - Hendrik Beling
- Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Thomas Galetin
- Department of Thoracic Surgery, Cologne-Merheim Hospital, Witten/Herdecke University Hospital, Cologne, Germany; Faculty of Medicine, Witten/Herdecke University, Witten, Germany
| | - Aris Koryllos
- Department of Thoracic Surgery, Cologne-Merheim Hospital, Witten/Herdecke University Hospital, Cologne, Germany; Faculty of Medicine, Witten/Herdecke University, Witten, Germany
| | - Erich Stoelben
- Department of Thoracic Surgery, Cologne-Merheim Hospital, Witten/Herdecke University Hospital, Cologne, Germany; Faculty of Medicine, University of Cologne, Cologne, Germany
| |
Collapse
|
20
|
Ying X, Fu W, Qi M, Zhou L, Li W, Su B. Ultrasensitive Electrochemical Vertical Flow Immunoassay for Rapid and Simultaneous Detection of Interleukin-6 and Procalcitonin. ACS Sens 2024; 9:6667-6674. [PMID: 39637373 DOI: 10.1021/acssensors.4c02257] [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] [Indexed: 12/07/2024]
Abstract
Rapid and multiplexed detection of biomarkers plays an indispensable role in disease diagnosis. Although paper-based lateral flow immunoassays have been widely used in this field, the speediness and throughput are still challenging issues. Herein, an electrochemical vertical flow immunoassay device (eVFID) is fabricated for rapid, ultrasensitive, and multiplexed detection of inflammatory biomarkers. Working electrodes with excellent electrochemical performance and permeability properties were directly fabricated on the nitrocellulose membrane to enable both the vertical flow of the sample solution and electrochemical detection. This vertical configuration can remarkably improve the speediness of the immunoassay and effectively inhibit the cross-talk reactions among immunomolecules, thus allowing rapid and simultaneous detection of multiplexed biomarkers. Furthermore, a signal amplification strategy based on horseradish peroxidase and tetramethylbenzidine was integrated into the eVFID to substantially increase the sensitivity of the electrochemical detection. A low limit of detection of 0.1 and 0.22 pg mL-1 was obtained for two low-abundance inflammatory biomarkers, interleukin-6 (IL-6) and procalcitonin (PCT), respectively. Finally, using a two-channel eVFID, simultaneous detection of IL-6 and PCT in human plasma samples was successfully realized within 5 min. We believe that the eVFID holds great promise for speedy and high-throughput biomarker detection at the point of care.
Collapse
Affiliation(s)
- Xudong Ying
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Wenxuan Fu
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Min Qi
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Lin Zhou
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Wei Li
- Department of Clinical Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Bin Su
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
21
|
Li KJ, Zhang ZY, Sulayman S, Shu Y, Wang K, Ababaike S, Zeng XY, Zhao ZL. Prognostic value of combined systemic inflammation response index and prognostic nutritional index in colorectal cancer patients. World J Gastrointest Surg 2024; 16:3794-3805. [PMID: 39734462 PMCID: PMC11650238 DOI: 10.4240/wjgs.v16.i12.3794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/05/2024] [Accepted: 10/22/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND The prognosis of colorectal cancer (CRC) patients is notably influenced by both inflammation and nutritional status. The prognostic nutritional index (PNI) and systemic inflammatory response index (SIRI) have been reported in prognostic studies of various tumors. However, the efficacy of the combination of the two in predicting the prognosis of CRC patients has not been studied. AIM To evaluate the effectiveness of PNI and SIRI in predicting the prognosis of patients with CRC. METHODS We retrospectively gathered data from 470 CRC patients who underwent feasible radical surgery at Xinjiang Cancer Hospital. The optimal cut-off values for SIRI and PNI, along with their predictive power for survival, were determined through area under the receiver operating characteristic curve using time-dependent receiver operating characteristic analysis. The Kaplan-Meier method and log-rank test were applied to assess prognostic impact, and a multifactorial Cox proportional hazards model was employed for analysis. Additionally, a new model, PSIRI, was developed and assessed for its survival prediction capability. RESULTS The optimal cutoff values for PNI and SIRI were determined to be 47.80 and 1.38, respectively. Based on these values, patients were categorized into high PNI and low PNI groups, as well as high SIRI and low SIRI groups. Significant differences in age, T stage, neutrophil to lymphocyte ratio (NLR), monocyte to lymphocyte ratio (MLR), and platelet-to-lymphocyte ratio (PLR) subgroups were observed between the PNI groups in the baseline profile. In the SIRI group, notable differences were found in gender, T stage, nerve invasion, intravascular tumor emboli, NLR, MLR, and PLR subgroups. Both low PNI and high SIRI were identified as independent risk factors for poor prognosis in CRC patients. When combined into the PSIRI model, it was shown that patients with a PSIRI ≤ 1 had a higher risk of death compared to those with a PSIRI of 2. CONCLUSION We assessed the impact of PNI and SIRI on the prognostic survival of CRC patients and developed a new model, PSIRI. This model demonstrated superior predictive accuracy, with a concordance index of 0.767.
Collapse
Affiliation(s)
- Ke-Jin Li
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Zi-Yi Zhang
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Subinur Sulayman
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Yin Shu
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Kuan Wang
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Saibihutula Ababaike
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Xiang-Yue Zeng
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Ze-Liang Zhao
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
22
|
Amer H, Kampan NC, Itsiopoulos C, Flanagan KL, Scott CL, Kartikasari AER, Plebanski M. Interleukin-6 Modulation in Ovarian Cancer Necessitates a Targeted Strategy: From the Approved to Emerging Therapies. Cancers (Basel) 2024; 16:4187. [PMID: 39766086 PMCID: PMC11674514 DOI: 10.3390/cancers16244187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Despite significant advances in treatments, ovarian cancer (OC) remains one of the most prevalent and lethal gynecological cancers in women. The frequent detection at the advanced stages has contributed to low survival rates, resistance to various treatments, and disease recurrence. Thus, a more effective approach is warranted to combat OC. The cytokine Interleukin-6 (IL6) has been implicated in various stages of OC development. High IL6 levels are also correlated with a lower survival rate in OC patients. In this current review, we summarized the pivotal roles of IL6 in OC, including the initiation, development, invasion, metastasis, and drug resistance mechanisms. This article systematically highlights how targeting IL6 improves OC outcomes by altering various cancer processes and reports the ongoing clinical trials that would further shape the IL6-based targeted therapies. This review also suggests how combining IL6-targeted therapies with other therapeutic strategies could further enhance their efficacy to combat OC.
Collapse
Affiliation(s)
- Hina Amer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia; (H.A.); (A.E.R.K.)
| | - Nirmala C. Kampan
- Department of Obstetrics and Gynecology, Faculty of Medicine, University Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Catherine Itsiopoulos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia; (H.A.); (A.E.R.K.)
| | - Katie L. Flanagan
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia; (H.A.); (A.E.R.K.)
- School of Medicine and School of Health Sciences, University of Tasmania, Launceston, TAS 7250, Australia
- Tasmanian Vaccine Trial Centre, Clifford Craig Foundation, Launceston General Hospital, Launceston, TAS 7250, Australia
| | - Clare L. Scott
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Faculty of Medicine, Dentistry, and Health Sciences, The University of Melbourne, Parkville, VIC 3052, Australia
- The Royal Women’s Hospital, Parkville, VIC 3052, Australia
| | - Apriliana E. R. Kartikasari
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia; (H.A.); (A.E.R.K.)
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia; (H.A.); (A.E.R.K.)
| |
Collapse
|
23
|
Kartikasari AER, Michel-Lara P, Exton H, Tekin-Sari K, Alnefai EMM, Mitchell A, Sanchez-Huertas C, Plebanski M. Circulating microRNAs as Diagnostic Biomarkers to Detect Specific Stages of Ovarian Cancer: A Comprehensive Meta-Analysis. Cancers (Basel) 2024; 16:4190. [PMID: 39766088 PMCID: PMC11674734 DOI: 10.3390/cancers16244190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Ovarian cancer (OC) is one of the most common gynecological cancers [...].
Collapse
Affiliation(s)
- Apriliana Ellya Ratna Kartikasari
- Cancer, Ageing, and Vaccine Research Group (CAVA), School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia (E.M.M.A.)
| | - Paul Michel-Lara
- Integrated Photonics and Applications Centre (InPAC), School of Engineering, RMIT University, Melbourne 3001, Australia
| | - Hayden Exton
- Cancer, Ageing, and Vaccine Research Group (CAVA), School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia (E.M.M.A.)
| | - Kaan Tekin-Sari
- Cancer, Ageing, and Vaccine Research Group (CAVA), School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia (E.M.M.A.)
| | - Ebtesam Motlaq M. Alnefai
- Cancer, Ageing, and Vaccine Research Group (CAVA), School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia (E.M.M.A.)
| | - Arnan Mitchell
- Integrated Photonics and Applications Centre (InPAC), School of Engineering, RMIT University, Melbourne 3001, Australia
| | - Cesar Sanchez-Huertas
- Integrated Photonics and Applications Centre (InPAC), School of Engineering, RMIT University, Melbourne 3001, Australia
| | - Magdalena Plebanski
- Cancer, Ageing, and Vaccine Research Group (CAVA), School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia (E.M.M.A.)
| |
Collapse
|
24
|
Sabra ASIM, Moselhy SNA, Eldin AKMZ. Systemic inflammatory indices as a non-invasive grading modality for endometriosis: a comparative study versus exploratory laparoscopy. REVISTA BRASILEIRA DE GINECOLOGIA E OBSTETRÍCIA 2024; 46:e-rbgo84. [PMID: 39669306 PMCID: PMC11637453 DOI: 10.61622/rbgo/2024rbgo84] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/05/2024] [Indexed: 12/14/2024] Open
Abstract
Objective Included evaluation of the possibility of using the systemic inflammatory indices for preoperative screening for the presence and severity of endometriosis (EM) in comparison to the findings of the exploratory laparoscopy. Methods 88 women with clinical manifestations suggestive of EM were evaluated clinically and by US and gave blood samples for estimation of serum cancer antigen-125 (CA125), platelet and total and differential leucocytic counts for calculation of inflammatory indices; the Systemic Immune-Inflammation index, the Systemic Inflammation Response Index (SIRI), the Neutrophil-Lymphocyte ratio (NLR), the Neutrophil-Monocyte ratio, the Neutrophil-Platelet ratio and the Platelet-Lymphocyte ratio. Then, patients were prepared to undergo laparoscopy for diagnosis and staging. Results Laparoscopy detected EM lesions in 63 patients; 27 of stage I-II and 36 of stage III-IV. Positive laparoscopy showed significant relation with US grading, high serum CA125 levels, platelet and inflammatory cell counts and indices. Statistical analyses defined high SIRI and NLR as the significant predictors for positive laparoscopy and high serum CA125 and NLR as the most significant predictors for severe EM (stage III-IV) on laparoscopy. Conclusion The intimate relation between EM and inflammation was reflected systematically as high levels of blood cellular components, but indices related to neutrophil especially NLR and SIRI showed highly significant relation to the presence and severity of EM and might be used as routine, cheap and non-invasive screening test before exploratory laparoscopy to guide the decision-making.
Collapse
Affiliation(s)
- Ahmed Sabra Ibrahim Mohammed Sabra
- Benha UniversityFaculty of MedicineDepartment of Obstetrics and GynecologyBenhaEgyptDepartment of Obstetrics and Gynecology, Faculty of Medicine, Benha University, Benha, Egypt.
| | - Shreen Naguib Aboelezz Moselhy
- Benha UniversityFaculty of MedicineDepartment of Obstetrics and GynecologyBenhaEgyptDepartment of Obstetrics and Gynecology, Faculty of Medicine, Benha University, Benha, Egypt.
| | - Ahmed Kasem Mohamed Zain Eldin
- Benha UniversityFaculty of MedicineDepartment of Obstetrics and GynecologyBenhaEgyptDepartment of Obstetrics and Gynecology, Faculty of Medicine, Benha University, Benha, Egypt.
| |
Collapse
|
25
|
Shirzad M, Daraei A, Najafzadehvarzi H, Farnoush N, Parsian H. Co-culture system of breast cancer and normal cells to investigate inflammation: using doxorubicin encapsulated in adipose-derived exosomes. Med Oncol 2024; 42:21. [PMID: 39630192 DOI: 10.1007/s12032-024-02568-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/11/2024] [Indexed: 01/23/2025]
Abstract
Doxorubicin (DOX) chemotherapy for breast cancer is an effective treatment option, but it also has disadvantages. Exosomes (EXOs) have safely and successfully transported DOX and reduced its adverse effects; however, its use is still being explored. In this study, a co-culture system of malignant and non-malignant breast cells was used to generate an in vitro model reflecting the in vivo cellular microenvironment, and the effects of this treatment were investigated by examining inflammatory genes. Extracellular matrices (EXOs) were extracted from mesenchymal stem cells derived from human adipose tissue by ultracentrifugation. Later, Western blotting, dynamic light scattering (DLS) and transmission electron microscopy methods were used to examine the properties of the EXO. DOX was encapsulated in the EXOs by sonication and the loading rate was measured by spectrophotometry. In the current study, a co-culture system was used to investigate the cytotoxic effects of free DOX and DOX encapsulated in EXOs (EXO-DOX) on various breast cell lines, including MCF-7, MCF-10A, MDA-MB-231, and A-MSC. Additionally, the expression levels of inflammatory cytokines (IL-1β, IL-6, IL-10, and TNF-α) were examined. Methylthiazolyldiphenyl-tetrazolium bromide assay demonstrated that free DOX showed the highest cytotoxicity against MCF-10A cells, followed by MCF-7 cells. Conversely, EXO-DOX indicated a greater effect on MCF-7 cells and had a lower IC50 compared to MDA-MB-231 cells. Free DOX significantly downregulated the expression of pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α), particularly in MCF-7 and MCF-10A cells, while concurrently upregulating IL-10 expression. EXO-DOX induced a more significant alteration in cytokine expression than the control and free DOX treatment groups. The co-culture system revealed a synergistic effect of free DOX on cancer cells while simultaneously mitigating the toxic effects of DOX on normal cells. This study suggests that EXO-DOX has promising potential as a targeted drug delivery system that could potentially improve therapeutic efficacy and minimize off-target toxicity.
Collapse
Affiliation(s)
- Moein Shirzad
- Student Research Committee, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Abdolreza Daraei
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Hossein Najafzadehvarzi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Pharmacology Department, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Nazila Farnoush
- Department of Surgery, Babol University of Medical Sciences, Babol, Iran
| | - Hadi Parsian
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
26
|
Catalano G, Alaimo L, Chatzipanagiotou OP, Ruzzenente A, Aucejo F, Marques HP, Lam V, Hugh T, Bhimani N, Maithel SK, Kitago M, Endo I, Martel G, Pulitano C, Shen F, Popescu I, Koerkamp BG, Bauer TW, Cauchy F, Poultsides GA, Weiss M, Gleisner A, Pawlik TM. Prognostic value of the advanced lung cancer inflammation index in intrahepatic cholangiocarcinoma. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108773. [PMID: 39447453 DOI: 10.1016/j.ejso.2024.108773] [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: 09/10/2024] [Revised: 10/08/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
INTRODUCTION The advanced lung cancer inflammation index (ALI), which combines inflammation and nutrition data, was recently proposed as a prognostic biomarker. We assessed the impact of ALI on overall survival (OS) among patients undergoing surgery for intrahepatic cholangiocarcinoma (ICC). METHODS Patients who underwent surgery for ICC were identified from an international cohort. ALI was calculated as body-mass index (BMI)∗albumin/neutrophil-to-lymphocyte ratio; patients were categorized into "low-" and "high-ALI" using log-rank statistics. The impact of ALI on OS was compared against other inflammatory markers (i.e., neutrophil-to-lymphocyte ratio [NLR], platelet-to-lymphocyte ratio [PLR], systemic immune inflammation index [SII = platelets∗NLR]) using Harrell's Concordance index (C-index) and the Akaike Information Criterion (AIC). To minimize intergroup differences, propensity score matching was employed. RESULTS Among 1045 patients, more than one-half of individuals underwent major hepatectomy (n = 582, 55.7 %), median tumor size was 5.5 cm (IQR, 3.8-7.8), and median ALI was 38.9 (IQR 26.5-57.2). On multivariate analysis, low ALI was an independent risk factor for worse OS (HR 1.21, 95 % CI 1.01-1.46; p = 0.04). Patients with low ALI had worse 5-year OS (36.9 % vs. 49.9 %; p < 0.001), which remained significant after PSM (36.9 % vs. 41.3 %; p = 0.039). ALI had a comparable discriminatory ability compared with NLR, PLR, and SII (C-index: 0.646 vs. 0.644 vs. 0.640 vs. 0.641, respectively), yet had a lower AIC (5475.31 vs. 5546.80 vs. 5550.45 vs. 5548.62, respectively) suggesting slightly better model fit and accuracy. CONCLUSIONS ALI was an independent predictor of OS among patients undergoing surgery for ICC. Nutritional and inflammatory markers should be incorporated into predictive models to improve prognostic stratification.
Collapse
Affiliation(s)
- Giovanni Catalano
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Surgery, University of Verona, Verona, Italy
| | - Laura Alaimo
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Surgery, University of Verona, Verona, Italy
| | | | | | - Federico Aucejo
- Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Cleveland, OH, USA
| | - Hugo P Marques
- Department of Surgery, Curry Cabral Hospital, Lisbon, Portugal
| | - Vincent Lam
- Department of Surgery, Westmead Hospital, Sydney, Australia
| | - Tom Hugh
- Department of Surgery, The University of Sydney, School of Medicine, Sydney, Australia
| | - Nazim Bhimani
- Department of Surgery, The University of Sydney, School of Medicine, Sydney, Australia
| | - Shishir K Maithel
- Division of Surgical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Minoru Kitago
- Department of Surgery, Keio University, Tokyo, Japan
| | - Itaru Endo
- Yokohama City University School of Medicine, Yokohama, Japan
| | | | - Carlo Pulitano
- Department of Surgery, Royal Prince Alfred Hospital, University of Sydney, Sydney, NSW, Australia
| | - Feng Shen
- Department of Surgery, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Irinel Popescu
- Department of Surgery, Fundeni Clinical Institute, Bucharest, Romania
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Todd W Bauer
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - François Cauchy
- Department of Hepatobiliopancreatic Surgery and Liver Transplantation, AP-HP, Beaujon Hospital, Clichy, France
| | | | - Matthew Weiss
- Department of Surgery, Cancer Institute, Northwell Health, NY, USA
| | - Ana Gleisner
- Department of Surgery, UC Denver, Denver, CO, USA
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
27
|
Lam KC, Goldszmid RS. Single-cell RNA flow cytometry to assess intratumoral production of cytokines/chemokines. Methods Cell Biol 2024; 191:221-246. [PMID: 39824558 DOI: 10.1016/bs.mcb.2024.10.013] [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] [Indexed: 01/20/2025]
Abstract
The tumor microenvironment (TME) consists of complex interactions between cellular and extracellular components, among which the immune system is known to play an integral role in disease progression and response to therapy. Cytokines and chemokines are cell signaling proteins used by immune cells to communicate with each other as well as with other cell types in the body. These proteins control systemic and local immune responses and levels of cytokines/chemokines in the TME have been associated with tumor outcomes. However, cytokines and chemokines have varied expression across cell types, tumors, and host conditions. Therefore, approaches to effectively study the production of these proteins at the single-cell level in the TME are needed to fully elucidate the mechanisms governing the anti-cancer immune response. Here, we detail a protocol to assess the production of cytokines/chemokines across leukocyte populations in mouse tumors using RNA flow cytometry. Importantly, this method can be adapted with minimal changes to study various mouse and human tumors, other RNA analytes, and non-tumor tissues.
Collapse
Affiliation(s)
- Khiem C Lam
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology (LICI), Center for Cancer Research (CCR), National Cancer Institute (NCI), Bethesda, MD, United States; Computational Biology, Bioinformatics and Genomics, Biological Sciences, University of Maryland, College Park, MD, United States
| | - Romina S Goldszmid
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology (LICI), Center for Cancer Research (CCR), National Cancer Institute (NCI), Bethesda, MD, United States.
| |
Collapse
|
28
|
Moralev A, Zenkova MA, Markov AV. Complex Inhibitory Activity of Pentacyclic Triterpenoids against Cutaneous Melanoma In Vitro and In Vivo: A Literature Review and Reconstruction of Their Melanoma-Related Protein Interactome. ACS Pharmacol Transl Sci 2024; 7:3358-3384. [PMID: 39539268 PMCID: PMC11555519 DOI: 10.1021/acsptsci.4c00422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/13/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
Pentacyclic triterpenoids (PTs) are a class of plant metabolites with a wide range of pharmacological activities, including strong antitumor potential against skin malignancies. By acting on multiple signaling pathways that control key cellular processes, PTs are able to exert complex effects on melanoma progression in vitro and in vivo. In this review, we have analyzed the works published in the past decade and devoted to the effects of PTs, both natural and semisynthetic, on cutaneous melanoma pathogenesis, including not only their direct action on melanoma cells but also their influence on the tumor microenvironment and abberant melanogenesis, often associated with melanoma aggressiveness. Special attention will be paid to the molecular basis of the pronounced antimelanoma potency of PTs, including a detailed consideration of the pathways sensitive to PTs in melanoma cells, as well as the reconstruction of the melanoma-related protein interactome of PTs using a network pharmacology approach based on previously published experimentally verified protein targets of PTs. The information collected on the primary targets of PTs was compiled in the Protein Interactome of PTs (PIPTs) database, freely available at http://www.pipts-db.ru/, which can be used to further optimize the mechanistic studies of PTs in the context of melanoma and other malignancies. By summarizing recent research findings, this review provides valuable information to scientists working in the fields related to the evaluation of melanoma pathogenesis and development of PTs-based drug candidates.
Collapse
Affiliation(s)
- Arseny
D. Moralev
- Institute of Chemical Biology and Fundamental
Medicine, Siberian Branch of the Russian
Academy of Sciences, 630090, Lavrent’ev avenue 8, Novosibirsk, Russia
| | - Marina A. Zenkova
- Institute of Chemical Biology and Fundamental
Medicine, Siberian Branch of the Russian
Academy of Sciences, 630090, Lavrent’ev avenue 8, Novosibirsk, Russia
| | - Andrey V. Markov
- Institute of Chemical Biology and Fundamental
Medicine, Siberian Branch of the Russian
Academy of Sciences, 630090, Lavrent’ev avenue 8, Novosibirsk, Russia
| |
Collapse
|
29
|
Shen Q, Murakami K, Sotov V, Butler M, Ohashi PS, Reedijk M. Inhibition of Notch enhances efficacy of immune checkpoint blockade in triple-negative breast cancer. SCIENCE ADVANCES 2024; 10:eado8275. [PMID: 39475614 PMCID: PMC11524187 DOI: 10.1126/sciadv.ado8275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024]
Abstract
Aberrant Notch, which is a defining feature of triple-negative breast cancer (TNBC) cells, regulates intercellular communication in the tumor immune microenvironment (TIME). This includes tumor-associated macrophage (TAM) recruitment through Notch-dependent cytokine secretion, contributing to an immunosuppressive TIME. Despite the low response rate of TNBC to immune checkpoint blockade (ICB), here, we report that inhibition of Notch-driven cytokine-mediated programs reduces TAMs and induces responsiveness to sequentially delivered ICB. This is characterized by the emergence of GrB+ cytotoxic T lymphocytes (CTLs) in the primary tumor. A more impressive effect of sequential treatment is observed in the lung where TAM depletion and increased CTLs are accompanied by near-complete abolition of metastases. This is due to (i) therapeutic reduction in Notch-dependent, prometastatic circulating factors released by the primary tumor, and (ii) elevated PD ligand 1 (PD-L1) in lung metastases, rendering them profoundly sensitive to ICB. These findings highlight the potential of combination cytokine inhibition and ICB as an immunotherapeutic strategy in TNBC.
Collapse
Affiliation(s)
- Qiang Shen
- Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| | - Kiichi Murakami
- Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| | - Valentin Sotov
- Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| | - Marcus Butler
- Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, Division of Medical Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Pamela S. Ohashi
- Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
- Department of Immunology, University of Toronto, Medical Sciences Building, 1 King’s College Circle, Room 7205, Toronto, Ontario M5S 1A8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto Medical Discovery Tower, MaRS Centre, 101 College Street, Room 15-701, Toronto, Ontario M5G 2M9, Canada
| | - Michael Reedijk
- Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
- Department of Medical Biophysics, University of Toronto, Toronto Medical Discovery Tower, MaRS Centre, 101 College Street, Room 15-701, Toronto, Ontario M5G 2M9, Canada
- Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Suite 8-411, Toronto, Ontario M5G 2M9, Canada
| |
Collapse
|
30
|
Chhipa AS, Boscaro V, Gallicchio M, Patel S. The curious case of type I interferon signaling in cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189204. [PMID: 39477031 DOI: 10.1016/j.bbcan.2024.189204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
Cytokines are the crucial signaling proteins that mediate the crosstalks between the cells of tumor microenvironment (TME). Interferon-1 (IFN-1) are the important cytokines that are widely known for their tumor suppressive roles comprising of cancer cell intrinsic and extrinsic mechanisms. Despite having known antitumor effects, IFN-1 are also reported to have tumor promoting functions under varying circumstances. This dichotomy in the functions of IFN-1 is largely attributed to the acute and chronic activation of IFN-1 signaling in TME. The chronic activation of IFN-1 signaling in tumor cells results in altered stimulation of downstream pathways that result in the expression of tumor promoting proteins, while the acute IFN-1 signaling activation maintains its tumor inhibiting functions. In the present review, we have discussed the anti- and pro-tumor actions of IFN-1 signaling under acute and chronic IFN-1 signaling activation. We have also discussed the downstream changes in signaling components that result in tumor supportive functions of a constitutive IFN-1 signaling. We have further discussed the possible strategies to overcome the detrimental effects of chronic IFN-1 pathway activation and to successfully employ IFN-1 for their beneficial anti-tumor effects.
Collapse
Affiliation(s)
- Abu Sufiyan Chhipa
- Department of Pharmacology, Institute of Pharmacy, Nirma University, 382481 Ahmedabad, India; Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Valentina Boscaro
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | | | - Snehal Patel
- Department of Pharmacology, Institute of Pharmacy, Nirma University, 382481 Ahmedabad, India.
| |
Collapse
|
31
|
Wang R, Liu J, Jiang B, Gao B, Luo H, Yang F, Ye Y, Chen Z, Liu H, Cui C, Xu K, Li B, Yang X. A single-cell perspective on immunotherapy for pancreatic cancer: from microenvironment analysis to therapeutic strategy innovation. Front Immunol 2024; 15:1454833. [PMID: 39539544 PMCID: PMC11557317 DOI: 10.3389/fimmu.2024.1454833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Pancreatic cancer remains one of the most lethal malignancies, with conventional treatment options providing limited efficacy. Recent advancements in immunotherapy have offered new hope, yet the unique tumor microenvironment (TME) of pancreatic cancer poses significant challenges to its successful application. This review explores the transformative impact of single-cell technology on the understanding and treatment of pancreatic cancer. By enabling high-resolution analysis of cellular heterogeneity within the TME, single-cell approaches have elucidated the complex interplay between various immune and tumor cell populations. These insights have led to the identification of predictive biomarkers and the development of innovative, personalized immunotherapeutic strategies. The review discusses the role of single-cell technology in dissecting the intricate immune landscape of pancreatic cancer, highlighting the discovery of T cell exhaustion profiles and macrophage polarization states that influence treatment response. Moreover, it outlines the potential of single-cell data in guiding the selection of immunotherapy drugs and optimizing treatment plans. The review also addresses the challenges and prospects of translating these single-cell-based innovations into clinical practice, emphasizing the need for interdisciplinary research and the integration of artificial intelligence to overcome current limitations. Ultimately, the review underscores the promise of single-cell technology in driving therapeutic strategy innovation and improving patient outcomes in the battle against pancreatic cancer.
Collapse
Affiliation(s)
- Rui Wang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- General Surgery Day Ward, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
| | - Jie Liu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Bo Jiang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Benjian Gao
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Honghao Luo
- Department of Radiology, Xichong People’s Hospital, Nanchong, China
| | - Fengyi Yang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuntao Ye
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zhuo Chen
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Hong Liu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Cheng Cui
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ke Xu
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Bo Li
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaoli Yang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
32
|
Tai YS, Leung JH, Wang SY, Leung HWC, Chan ALF. Association of Granulocyte Colony-Stimulating Factor Treatment with Risk of Brain Metastasis in Advanced Stage Breast Cancer. Int J Mol Sci 2024; 25:10756. [PMID: 39409083 PMCID: PMC11477282 DOI: 10.3390/ijms251910756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
The routine use of granulocyte colony-stimulating factor (GCSF) is not recommended for the prevention or treatment of chemotherapy-induced neutropenia or febrile neutropenia because risks associated with certain types of cancers, distant organ metastases, and primary tumor growth cannot be excluded. We examined the association between GCSF use and the incidence of brain metastasis (BM), as well as BM-free survival (BMFS). This retrospective cohort study included 121 stage IV breast cancer patients without confirmed BM at the time of diagnosis and who received at least one course of systematic chemotherapy or target therapy at a tertiary teaching hospital between 1 January 2014 and 31 December 2022. The effect of GCSF use on BM was assessed with other confounding factors in Cox regression analyses. In this retrospective cohort, patients who received GCSF treatment had a significantly higher incidence of BM than those who did not (34.9% vs. 13.8%, p = 0.011). Univariate Cox regression analysis showed that GCSF use, menopause status, hormone treatment, HER2 treatment, cumulative dosage, dosage density, and neutropenia were independent risk factors for BMFS (p < 0.05). GCSF users had a higher risk of BM (adjusted HR: 2.538; 95% CI: 1.127-5.716, p = 0.025) than nonusers. BM risk was significantly associated with those with neutropenia (RR: 1.84, 95% CI: 1.21, 2.80) but not with those without neutropenia (RR: 0.59, 95% CI: 0.41-0.84, Interaction p-value < 0.05). The higher the dose density of GCSF, the higher the risk compared with those who do not use GCSF (p for trend < 0.01). These preliminary results suggest that GCSF is associated with BM in patients with stage IV breast cancer who did not have BM at initial diagnosis. Further comprehensively designed large-scale observational studies are needed to confirm our preliminary results.
Collapse
Affiliation(s)
- Yun-Sheng Tai
- Department of Breast Surgery, An-Nan Hospital, China Medical University, Tainan 709, Taiwan;
| | - John Hang Leung
- Department of Obstetrics and Gynecology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi 600, Taiwan;
| | - Shyh-Yau Wang
- Department of Radiology, An-Nan Hospital, China Medical University, Tainan 709, Taiwan;
| | - Henry W. C. Leung
- Department of Radiation Oncology, An-Nan Hospital, China Medical University, Tainan 709, Taiwan
| | - Agnes L. F. Chan
- Department of Pharmacy, Kaohsiung Show Chwan Memorial Hospital, Kaohsiung 821, Taiwan
| |
Collapse
|
33
|
Appel K, Rose T, Zimmermann C, Günnewich N. In Vitro Anti-inflammatory Effects of Larch Turpentine, Turpentine Oil, Eucalyptus Oil, and Their Mixture as Contained in a Marketed Ointment. PLANTA MEDICA 2024; 90:1023-1029. [PMID: 39260387 PMCID: PMC11614573 DOI: 10.1055/a-2388-7527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 08/01/2024] [Indexed: 09/13/2024]
Abstract
An ointment containing larch turpentine, turpentine oil, and eucalyptus oil has been used for almost a century for the symptomatic treatment of mild, localized, purulent inflammations of the skin. Its clinical efficacy in the treatment of skin infections has been shown in clinical trials, but the mode of action of the active ingredients on inflammation is not known. We studied the anti-inflammatory properties of the active ingredients of the ointment and their mixture in a human monocyte cell model, in which the cells were stimulated with lipopolysaccharide and incubated with the test substances. The cytotoxic threshold of each test substance and the mixture was identified using the alamarBlue assay, and their anti-inflammatory activity was assessed by measuring the release of interleukins IL-1β, IL-6, IL-8, monocyte chemoattractant protein-1, prostaglandin E2, and TNF-α. Cell toxicity was observed at a mixture concentration of 10 µg/mL. All immunological assays were carried out at nontoxic concentrations. Larch turpentine decreased IL-1β, monocyte chemoattractant protein-1, and prostaglandin E2 release at a concentration of 3.9 µg/mL and TNF-α at concentrations > 1.95 µg/mL, whereas eucalyptus oil and turpentine oil had no relevant inhibitory effects. The mixture dose-dependently inhibited IL-1β, IL-6, monocyte chemoattractant protein-1, prostaglandin E2, and TNF-α release at concentrations > 1 µg/mL. IL-8 release was only marginally affected. The anti-inflammatory activity of the herbal ingredients and their mixture was confirmed in this model. This effect seems to be mediated mainly by larch turpentine, with turpentine oil and eucalyptus oil exerting an additive or possibly synergistic function.
Collapse
Affiliation(s)
- Kurt Appel
- VivaCell Biotechnology GmbH, Denzlingen, Germany
| | | | | | | |
Collapse
|
34
|
Ramírez-Pacheco A, Moreno-Guerrero SS, Rocha-Ramírez LM, Hernández-Pliego G, Escobar-Sánchez MA, Reyes-López A, Sienra-Monge JJL, Juárez-Villegas LE. Role of Genetic Polymorphisms -238 G>A and -308 G>A, and Serum TNF-α Levels in a Cohort of Mexican Pediatric Neuroblastoma Patients: Preliminary Study. Int J Mol Sci 2024; 25:10590. [PMID: 39408920 PMCID: PMC11477149 DOI: 10.3390/ijms251910590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/05/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
The results of in vitro and in vivo studies have shown the pro-tumor effects of TNF-α, and this cytokine's increased expression is associated with poor prognosis in patients with some types of cancer. Our study objective was to evaluate the possible association of TNF-α genetic polymorphisms and serum levels with susceptibility and prognosis in a cohort of Mexican patients with NB. We performed PCR-RFLP and ELISA methods to analyze the genetics of these SNPs and determine serum concentrations, respectively. The distribution of the -308 G>A and -238 G>A polymorphisms TNFα genotypes was considerably different between patients with NB and the control group. The SNP rs1800629 GG/GA genotypes were associated with a decreased risk of NB (OR = 0.1, 95% CI = 0.03-0.393, p = 0.001) compared with the AA genotype, which was associated with susceptibility to NB (OR = 2.89, 95% CI = 1.45-5.76, p = 0.003) and related to unfavorable histology and high-risk NB. The rs361525 polymorphism GG genotype was associated with a lower risk of developing NB compared with the GA and AA genotypes (OR = 0.2, 95% CI = 0.068-0.63, p = 0.006). Circulating TNF-α serum concentrations were significantly different (p < 0.001) between patients with NB and healthy controls; however, we found no relationship between the analyzed TNF-α serum levels and SNP genotypes. We found associations between the rs1800629AA genotype and lower event-free survival (p = 0.026); SNP rs361525 and TNF-α levels were not associated with survival in patients with NB. Our results suggest the TNF-α SNP rs1800629 as a probable factor of NB susceptibility. The -308 G/A polymorphism AA genotype has a probable role in promoting NB development and poor prognosis associated with unfavorable histology, high-risk tumors, and lower EFS in Mexican patients with NB. It should be noted that it is important to conduct research on a larger scale, through inter-institutional studies, to further evaluate the contribution of TNF-α genetic polymorphisms to the risk and prognosis of NB.
Collapse
Affiliation(s)
- Arturo Ramírez-Pacheco
- Departamento de Hemato-Oncología, Hospital Infantil de México Federico Gómez, Dr. Márquez No. 162, Col Doctores, Ciudad de México 06720, Mexico; (A.R.-P.); (S.S.M.-G.); (G.H.-P.)
| | - Silvia Selene Moreno-Guerrero
- Departamento de Hemato-Oncología, Hospital Infantil de México Federico Gómez, Dr. Márquez No. 162, Col Doctores, Ciudad de México 06720, Mexico; (A.R.-P.); (S.S.M.-G.); (G.H.-P.)
| | - Luz María Rocha-Ramírez
- Unidad de Investigación en Enfermedades Infecciosas, Hospital Infantil de México Federico Gómez, Dr. Márquez No. 162, Col Doctores, Ciudad de México 06720, Mexico
| | - Gabriela Hernández-Pliego
- Departamento de Hemato-Oncología, Hospital Infantil de México Federico Gómez, Dr. Márquez No. 162, Col Doctores, Ciudad de México 06720, Mexico; (A.R.-P.); (S.S.M.-G.); (G.H.-P.)
| | - María Argelia Escobar-Sánchez
- Departamento de Patología Clínica y Experimental, Hospital Infantil de México Federico Gómez, Dr. Márquez No. 162, Col Doctores, Ciudad de México 06720, Mexico;
| | - Alfonso Reyes-López
- Centro de Estudios Económicos y Sociales en Salud, Hospital Infantil de México Federico Gómez, Dr. Márquez No. 162, Col Doctores, Ciudad de México 06720, Mexico;
| | - Juan José Luis Sienra-Monge
- Subdirección de Pediatría Ambulatoria, Hospital Infantil de México Federico Gómez, Dr. Márquez No. 162, Col Doctores, Ciudad de México 06720, Mexico;
| | - Luis Enrique Juárez-Villegas
- Departamento de Hemato-Oncología, Hospital Infantil de México Federico Gómez, Dr. Márquez No. 162, Col Doctores, Ciudad de México 06720, Mexico; (A.R.-P.); (S.S.M.-G.); (G.H.-P.)
| |
Collapse
|
35
|
Meidenbauer J, Wachter M, Schulz SR, Mostafa N, Zülch L, Frey B, Fietkau R, Gaipl US, Jost T. Inhibition of ATM or ATR in combination with hypo-fractionated radiotherapy leads to a different immunophenotype on transcript and protein level in HNSCC. Front Oncol 2024; 14:1460150. [PMID: 39411143 PMCID: PMC11473424 DOI: 10.3389/fonc.2024.1460150] [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: 07/05/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024] Open
Abstract
Background The treatment of head and neck tumors remains a challenge due to their reduced radiosensitivity. Small molecule kinase inhibitors (smKI) that inhibit the DNA damage response, may increase the radiosensitivity of tumor cells. However, little is known about how the immunophenotype of the tumor cells is modulated thereby. Therefore, we investigated whether the combination of ATM or ATR inhibitors with hypo-fractionated radiotherapy (RT) has a different impact on the expression of immune checkpoint markers (extrinsic), the release of cytokines or the transcriptome (intrinsic) of head and neck squamous cell carcinoma (HNSCC) cells. Methods The toxic and immunogenic effects of the smKI AZD0156 (ATMi) and VE-822 (ATRi) in combination with a hypo-fractionated scheme of 2x5Gy RT on HPV-negative (HSC4, Cal-33) and HPV-positive (UM-SCC-47, UD-SCC-2) HNSCC cell lines were analyzed as follows: cell death (necrosis, apoptosis; detected by AnxV/PI), expression of immunostimulatory (ICOS-L, OX40-L, TNFSFR9, CD70) and immunosuppressive (PD-L1, PD-L2, HVEM) checkpoint marker using flow cytometry; the release of cytokines using multiplex ELISA and the gene expression of Cal-33 on mRNA level 48 h post-RT. Results Cell death was mainly induced by the combination of RT with both inhibitors, but stronger with ATRi. Further, the immune phenotype of cancer cells, not dying from combination therapy itself, is altered predominantly by RT+ATRi in an immune-stimulatory manner by the up-regulation of ICOS-L. However, the analysis of secreted cytokines after treatment of HNSCC cell lines revealed an ambivalent influence of both inhibitors, as we observed the intensified secretion of IL-6 and IL-8 after RT+ATRi. These findings were confirmed by RNAseq analysis and further the stronger immune-suppressive character of RT+ATMi was enlightened. We detected the down-regulation of a central protein of cytoplasmatic sensing pathways of nucleic acids, RIG-1, and found one immune-suppressive target, EDIL3, strongly up-regulated by RT+ATMi. Conclusion Independent of a restrictive toxicity, the combination of RT + either ATMi or ATRi leads to comprehensive and immune-modulating alterations in HNSCC. This includes pro-inflammatory signaling induced by RT + ATRi but also anti-inflammatory signals. These findings were confirmed by RNAseq analysis, which further highlighted the immune-suppressive nature of RT + ATMi.
Collapse
Affiliation(s)
- Julia Meidenbauer
- Translational Radiobiology, Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (EMN), Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Wachter
- Translational Radiobiology, Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (EMN), Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian R. Schulz
- Division of Molecular Immunology, Internal Medicine III, University Hospital Erlangen, Nikolaus-Fiebiger Center, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Nada Mostafa
- Translational Radiobiology, Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (EMN), Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lilli Zülch
- Translational Radiobiology, Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (EMN), Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Benjamin Frey
- Translational Radiobiology, Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (EMN), Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Uniklinikum Erlangen, Erlangen, Germany
- FAU Profile Center Immunomedicine Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (EMN), Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Uniklinikum Erlangen, Erlangen, Germany
- FAU Profile Center Immunomedicine Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Udo S. Gaipl
- Translational Radiobiology, Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (EMN), Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Uniklinikum Erlangen, Erlangen, Germany
- FAU Profile Center Immunomedicine Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tina Jost
- Translational Radiobiology, Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (EMN), Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
36
|
Jaiswal A, Shrivastav S, Kushwaha HR, Chaturvedi R, Singh RP. Oncogenic potential of SARS-CoV-2-targeting hallmarks of cancer pathways. Cell Commun Signal 2024; 22:447. [PMID: 39327555 PMCID: PMC11426004 DOI: 10.1186/s12964-024-01818-0] [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: 07/01/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
The 2019 outbreak of SARS-CoV-2 has caused a major worldwide health crisis with high rates of morbidity and death. Interestingly, it has also been linked to cancer, which begs the issue of whether it plays a role in carcinogenesis. Recent studies have revealed various mechanisms by which SARS-CoV-2 can influence oncogenic pathways, potentially promoting cancer development. The virus encodes several proteins that alter key signaling pathways associated with cancer hallmarks. Unlike classical oncogenic viruses, which transform cells through viral oncogenes or by activating host oncogenes, SARS-CoV-2 appears to promote tumorigenesis by inhibiting tumor suppressor genes and pathways while activating survival, proliferation, and inflammation-associated signaling cascades. Bioinformatic analyses and experimental studies have identified numerous interactions between SARS-CoV-2 proteins and cellular components involved in cancer-related processes. This review explores the intricate relationship between SARS-CoV-2 infection and cancer, focusing on the regulation of key hallmarks driving initiation, promotion and progression of cancer by viral proteins. By elucidating the underlying mechanisms driving cellular transformation, the potential of SARS-CoV-2 as an oncovirus is highlighted. Comprehending these interplays is essential to enhance our understanding of COVID-19 and cancer biology and further formulating strategies to alleviate SARS-CoV-2 influence on cancer consequences.
Collapse
Affiliation(s)
- Aishwarya Jaiswal
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Sanah Shrivastav
- SRM Institute of Science and Technology, Delhi-NCR Campus, Ghaziabad, Uttar Pradesh, India
| | - Hemant R Kushwaha
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Rupesh Chaturvedi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Rana P Singh
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India.
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
37
|
Maier JA, Castiglioni S, Petrelli A, Cannatelli R, Ferretti F, Pellegrino G, Sarzi Puttini P, Fiorina P, Ardizzone S. Immune-Mediated Inflammatory Diseases and Cancer - a dangerous liaison. Front Immunol 2024; 15:1436581. [PMID: 39359726 PMCID: PMC11445042 DOI: 10.3389/fimmu.2024.1436581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
Patients with Immune-Mediated Inflammatory Diseases (IMIDs) are known to have an elevated risk of developing cancer, but the exact causative factors remain subject to ongoing debate. This narrative review aims to present the available evidence concerning the intricate relationship between these two conditions. Environmental influences and genetic predisposition lead to a dysregulated immune response resulting in chronic inflammation, which is crucial in the pathogenesis of IMIDs and oncogenic processes. Mechanisms such as the inflammatory microenvironment, aberrant intercellular communication due to abnormal cytokine levels, excessive reparative responses, and pathological angiogenesis are involved. The chronic immunosuppression resulting from IMIDs treatments further adds to the complexity of the pathogenic scenario. In conclusion, this review highlights critical gaps in the current literature, suggesting potential avenues for future research. The intricate interplay between IMIDs and cancer necessitates more investigation to deepen our understanding and improve patient management.
Collapse
Affiliation(s)
- Jeanette A Maier
- Department of Biomedical and Clinical Sciences, Università di Milano, Milano, Italy
| | - Sara Castiglioni
- Department of Biomedical and Clinical Sciences, Università di Milano, Milano, Italy
| | - Alessandra Petrelli
- Department of Clinical Sciences and Community Health, University of Milan, Milano, Italy
| | | | | | | | - Piercarlo Sarzi Puttini
- Department of Biomedical and Clinical Sciences, Università di Milano, Milano, Italy
- IRCCS Ospedale Galeazzi-Sant'Ambrogio, Milano, Italy
| | - Paolo Fiorina
- Department of Biomedical and Clinical Sciences, Università di Milano, Milano, Italy
| | - Sandro Ardizzone
- Gastroenterology Unit, ASST Fatebenefratelli-Sacco, Milano, Italy
| |
Collapse
|
38
|
Zhang Y, Ding X, Zhang X, Li Y, Xu R, Li HJ, Zuo D, Chen G. Unveiling the contribution of tumor-associated macrophages in driving epithelial-mesenchymal transition: a review of mechanisms and therapeutic Strategies. Front Pharmacol 2024; 15:1404687. [PMID: 39286635 PMCID: PMC11402718 DOI: 10.3389/fphar.2024.1404687] [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: 03/21/2024] [Accepted: 08/15/2024] [Indexed: 09/19/2024] Open
Abstract
Tumor-associated macrophages (TAMs), fundamental constituents of the tumor microenvironment (TME), significantly influence cancer development, primarily by promoting epithelial-mesenchymal transition (EMT). EMT endows cancer cells with increased motility, invasiveness, and resistance to therapies, marking a pivotal juncture in cancer progression. The review begins with a detailed exposition on the origins of TAMs and their functional heterogeneity, providing a foundational understanding of TAM characteristics. Next, it delves into the specific molecular mechanisms through which TAMs induce EMT, including cytokines, chemokines and stromal cross-talking. Following this, the review explores TAM-induced EMT features in select cancer types with notable EMT characteristics, highlighting recent insights and the impact of TAMs on cancer progression. Finally, the review concludes with a discussion of potential therapeutic targets and strategies aimed at mitigating TAM infiltration and disrupting the EMT signaling network, thereby underscoring the potential of emerging treatments to combat TAM-mediated EMT in cancer. This comprehensive analysis reaffirms the necessity for continued exploration into TAMs' regulatory roles within cancer biology to refine therapeutic approaches and improve patient outcomes.
Collapse
Affiliation(s)
- Yijia Zhang
- Department of Pharmacy, Taizhou Second People's Hospital (Mental Health Center affiliated to Taizhou University School of Medicine), Taizhou University, Taizhou, Zhejiang, China
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaofei Ding
- Department of Pharmacology, Taizhou University, Taizhou, Zhejiang, China
| | - Xue Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Ye Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Rui Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Hai-Jun Li
- Department of Pharmacy, Taizhou Second People's Hospital (Mental Health Center affiliated to Taizhou University School of Medicine), Taizhou University, Taizhou, Zhejiang, China
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Guang Chen
- Department of Pharmacy, Taizhou Second People's Hospital (Mental Health Center affiliated to Taizhou University School of Medicine), Taizhou University, Taizhou, Zhejiang, China
- Department of Pharmacology, Taizhou University, Taizhou, Zhejiang, China
| |
Collapse
|
39
|
Jagadeesan D, Sathasivam KV, Fuloria NK, Balakrishnan V, Khor GH, Ravichandran M, Solyappan M, Fuloria S, Gupta G, Ahlawat A, Yadav G, Kaur P, Husseen B. Comprehensive insights into oral squamous cell carcinoma: Diagnosis, pathogenesis, and therapeutic advances. Pathol Res Pract 2024; 261:155489. [PMID: 39111016 DOI: 10.1016/j.prp.2024.155489] [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: 06/11/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 08/18/2024]
Abstract
Oral squamous cell carcinoma (OSCC) is considered the most common type of head and neck squamous cell carcinoma (HNSCC) as it holds 90 % of HNSCC cases that arise from multiple locations in the oral cavity. The last three decades witnessed little progress in the diagnosis and treatment of OSCC the aggressive tumor. However, in-depth knowledge about OSCC's pathogenesis, staging & grading, hallmarks, and causative factors is a prime requirement in advanced diagnosis and treatment for OSCC patients. Therefore present review was intended to comprehend the OSCCs' prevalence, staging & grading, molecular pathogenesis including premalignant stages, various hallmarks, etiology, diagnostic methods, treatment (including FDA-approved drugs with the mechanism of action and side effects), and theranostic agents. The current review updates that for a better understanding of OSCC progress tumor-promoting inflammation, sustained proliferative signaling, and growth-suppressive signals/apoptosis capacity evasion are the three most important hallmarks to be considered. This review suggests that among all the etiology factors the consumption of tobacco is the major contributor to the high incidence rate of OSCC. In OSCC diagnosis biopsy is considered the gold standard, however, toluidine blue staining is the easiest and non-invasive method with high accuracy. Although there are various therapeutic agents available for cancer treatment, however, a few only are approved by the FDA specifically for OSCC treatment. The present review recommends that among all available OSCC treatments, the antibody-based CAR-NK is a promising therapeutic approach for future cancer treatment. Presently review also suggests that theranostics have boosted the advancement of cancer diagnosis and treatment, however, additional work is required to refine the role of theranostics in combination with different modalities in cancer treatment.
Collapse
Affiliation(s)
- Dharshini Jagadeesan
- Department of Biotechnology, Faculty of Applied Sciences, AIMST University, Bedong, Kedah, Malaysia
| | - Kathiresan V Sathasivam
- Department of Biotechnology, Faculty of Applied Sciences, AIMST University, Bedong, Kedah, Malaysia
| | | | - Venugopal Balakrishnan
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia,11800 USM, Pulau Pinang, Malaysia
| | - Goot Heah Khor
- Centre of Preclinical Science Studies, Faculty of Dentistry, Universiti Teknologi MARA, Sungai Buloh Campus, Jalan Hospital, SungaiBuloh, Selangor 47000, Malaysia; Oral and Maxillofacial Cancer Research Group, Faculty of Dentistry, Universiti Teknologi MARA, Sungai Buloh Campus, Jalan Hospital, Sungai Buloh, Selangor 47000, Malaysia
| | - Manickam Ravichandran
- Department of Biotechnology, Faculty of Applied Sciences, AIMST University, Bedong, Kedah, Malaysia
| | - Maheswaran Solyappan
- Department of Biotechnology, Faculty of Applied Sciences, AIMST University, Bedong, Kedah, Malaysia
| | | | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Punjab, India; Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Abhilasha Ahlawat
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Geeta Yadav
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - Pandeep Kaur
- National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Beneen Husseen
- Medical Laboratory Technique College, the Islamic University, Najaf, Iraq; Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
| |
Collapse
|
40
|
Knepp B, Navi BB, Rodriguez F, DeAngelis LM, Elkind MSV, Iadecola C, Sherman CP, Tagawa ST, Saxena A, Ocean AJ, Hull H, Jickling G, Sharp FR, Ander BP, Stamova B. Ischemic Stroke with Comorbid Cancer Has Specific miRNA-mRNA Networks in Blood That Vary by Ischemic Stroke Mechanism. Ann Neurol 2024; 96:565-581. [PMID: 38874304 PMCID: PMC11849972 DOI: 10.1002/ana.26997] [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: 10/20/2023] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/15/2024]
Abstract
OBJECTIVE Approximately half of ischemic strokes (IS) in cancer patients are cryptogenic, with many presumed cardioembolic. We evaluated whether there were specific miRNA and mRNA transcriptome architectures in peripheral blood of IS patients with and without comorbid cancer, and between cardioembolic versus noncardioembolic IS etiologies in comorbid cancer. METHODS We studied patients with cancer and IS (CS; n = 42), stroke only (SO; n = 41), and cancer only (n = 28), and vascular risk factor-matched controls (n = 30). mRNA-Seq and miRNA-Seq data, analyzed with linear regression models, identified differentially expressed genes in CS versus SO and in cardioembolic versus noncardioembolic CS, and miRNA-mRNA regulatory pairs. Network-level analyses identified stroke etiology-specific responses in CS. RESULTS A total of 2,085 mRNAs and 31 miRNAs were differentially expressed between CS and SO. In CS, 122 and 35 miRNA-mRNA regulatory pairs, and 5 and 3 coexpressed gene modules, were associated with cardioembolic and noncardioembolic CS, respectively. Complement, growth factor, and immune/inflammatory pathways showed differences between IS etiologies in CS. A 15-gene biomarker panel assembled from a derivation cohort (n = 50) correctly classified 81% of CS and 71% of SO participants in a validation cohort (n = 33). Another 15-gene panel correctly identified etiologies for 13 of 13 CS-cardioembolic and 11 of 11 CS-noncardioembolic participants upon cross-validation; 11 of 16 CS-cryptogenic participants were predicted cardioembolic. INTERPRETATION We discovered unique mRNA and miRNA transcriptome architecture in CS and SO, and in CS with different IS etiologies. Cardioembolic and noncardioembolic etiologies in CS showed unique coexpression networks and potential master regulators. These may help distinguish CS from SO and identify IS etiology in cryptogenic CS patients. ANN NEUROL 2024;96:565-581.
Collapse
Affiliation(s)
- Bodie Knepp
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Babak B Navi
- Clinical and Translational Neuroscience Unit, Department of Neurology, Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, USA
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fernando Rodriguez
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Lisa M DeAngelis
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mitchell S V Elkind
- Department of Neurology, Vagelos College of Physicians and Surgeons and Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Costantino Iadecola
- Clinical and Translational Neuroscience Unit, Department of Neurology, Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, USA
| | - Carla P Sherman
- Clinical and Translational Neuroscience Unit, Department of Neurology, Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, USA
| | - Scott T Tagawa
- Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ashish Saxena
- Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Allyson J Ocean
- Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Heather Hull
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Glen Jickling
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Division of Neurology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Frank R Sharp
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Bradley P Ander
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Boryana Stamova
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
41
|
Talukdar J, Malik A, Kataki K, Choudhury BN, Baruah MN, Bhattacharyya M, Sarma MP, Bhattacharjee M, Basak M, Kashyap MP, Bhattacharjee S, Ali E, Keppen C, Kalita S, Kalita MJ, Das PP, Hazarika G, Deka AJ, Dutta K, Idris MG, Akhtar S, Medhi S. Expression of Interleukin-8, Interleukin-12 and Interleukin-13 in Esophageal Squamous Cell Carcinoma: Biomarker Potentiality and Prognostic Significance. J Gastrointest Cancer 2024; 55:1239-1255. [PMID: 38910194 DOI: 10.1007/s12029-024-01063-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2024] [Indexed: 06/25/2024]
Abstract
PURPOSE Interleukin-8 (IL8), Interleukin-12 (IL12) and Interleukin-13 (IL13) are cytokines that play regulatory role in cancer pathogenesis. We analysed their expression profile to evaluate as molecular biomarkers of esophageal squamous cell carcinoma (ESCC) and their association with different parameters and patient survival. METHODS Expression analysis was performed by Real time quantitative polymerase chain reaction and receiver operating characteristic (ROC) curve analysis was done. The expression profiles were associated with different clinicopathological and dietary factors. Survival and hazard analysis were also performed. RESULTS IL8 expression showed upregulation in tissue (p = 0.000) and blood samples (p = 0.481), IL12 expression showed downregulation in tissue samples (p = 0.064) and upregulation in blood samples (p = 0.689) and IL13 expression showed upregulation in tissue (p = 0.000) and blood samples (p = 0.006). IL13 expression in tissue showed the highest area under the curve (AUC) value (0.773) for ESCC diagnosis, followed by IL8 expression in tissue (0.704) and IL13 expression in blood (0.643). This study also reveals the correlation of studied cytokines in tissue and blood level. Different clinicopathological and dietary factors showed significant association (p < 0.05) with IL8, IL12 and IL13 expression and with survival of ESCC patients. IL8 expression in blood and IL12 expression in tissue and blood showed significant association (p < 0.05) with patient survival. CONCLUSION Altered expression of IL8, IL12 and IL13 may be associated with ESCC progression. Overexpression of IL8 and IL13 in tissue samples may be potential biomarkers for ESCC screening. Additionally, both survival and hazard analysis data indicate the effects of different parameters on the prognosis of ESCC patients.
Collapse
Affiliation(s)
- Jayasree Talukdar
- Department of Bioengineering and Technology, Gauhati University, Gawahati, Assam, India
| | - Abdul Malik
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Kangkana Kataki
- Department of Bioengineering and Technology, Gauhati University, Gawahati, Assam, India
| | | | - Munindra Narayan Baruah
- Department of Head and Neck Oncology, North East Cancer Hospital and Research Institute, Jorabat, Assam, India
| | - Mallika Bhattacharyya
- Department of Gastroentrology, Gauhati Medical College and Hospital, Guwahati, Assam, India
| | - Manash Pratim Sarma
- Program of Biotechnology, Faculty of Science, Assam down town University, Guwahati, Assam, India
| | - Minakshi Bhattacharjee
- Program of Biotechnology, Faculty of Science, Assam down town University, Guwahati, Assam, India
| | - Mrinmoy Basak
- Faculty of Pharmaceutical Sciences, Assam down town University, Guwahati, Assam, India
| | - Manash Pratim Kashyap
- Program of Statistics, Faculty of Science, Assam down town University, Guwahati, Assam, India
| | | | - Eyashin Ali
- Department of Bioengineering and Technology, Gauhati University, Gawahati, Assam, India
| | - Chenole Keppen
- Department of Bioengineering and Technology, Gauhati University, Gawahati, Assam, India
| | - Simanta Kalita
- Department of Bioengineering and Technology, Gauhati University, Gawahati, Assam, India
- Multidisciplinary Research Unit, Diphu Medical College and Hospital, Karbi Anglong, Assam, India
| | - Manash Jyoti Kalita
- Department of Bioengineering and Technology, Gauhati University, Gawahati, Assam, India
| | - Partha Pratim Das
- Multidisciplinary Research Unit, Fakhruddin Ali Ahmed Medical College and Hospital, Barpeta, Assam, India
| | - Gautam Hazarika
- Department of Bioengineering and Technology, Gauhati University, Gawahati, Assam, India
| | - Ankur Jyoti Deka
- Department of Bioengineering and Technology, Gauhati University, Gawahati, Assam, India
| | - Kalpajit Dutta
- Department of Bioengineering and Technology, Gauhati University, Gawahati, Assam, India
| | | | - Suhail Akhtar
- A. T. Still University of Health Sciences, Kirksville, MO, USA
| | - Subhash Medhi
- Department of Bioengineering and Technology, Gauhati University, Gawahati, Assam, India.
| |
Collapse
|
42
|
Bujnakova Mlynarcikova A, Scsukova S. Evaluation of effects of bisphenol analogs AF, S, and F on viability, proliferation, production of selected cancer-related factors, and expression of selected transcripts in Caov-3 human ovarian epithelial cell line. Food Chem Toxicol 2024; 191:114889. [PMID: 39059691 DOI: 10.1016/j.fct.2024.114889] [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: 10/09/2023] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
Bisphenol A (BPA) has been a substantial additive in plastics until the reports on its adverse effects have led to its restrictions and replacement. Monitoring studies document the increasing occurrence of bisphenol analogs, however, data on their effects and risks is still insufficient. Based on the indications that BPA might contribute to ovarian cancer pathogenesis, we examined effects of the analogs AF (BPAF), S (BPS) and F (BPF) (10-9-10-4 M) on the Caov-3 epithelial cancer cells, including the impact on cell viability, proliferation, oxidative stress, and production and expression of several factors and genes related to ovarian cancer. At environmentally relevant doses, bisphenols did not exert significant effects. At the highest concentration, BPAF caused varied alterations, including decreased cell viability and proliferation, caspase activation, down-regulation of PCNA and BIRC5, elevation of IL8, VEGFA, MYC, PTGS2 and ABCB1 expressions. Only BPA (10-4 M) increased IL-6, IL-8 and VEGFA output by the Caov-3 cells. Each bisphenol induced generation of reactive oxygen species and decreased superoxide dismutase activity at the highest concentration. Although the effects were observed only in the supraphysiological doses, the results indicate that certain bisphenol analogs might affect several ovarian cancer cell characteristics and merit further investigation.
Collapse
Affiliation(s)
- Alzbeta Bujnakova Mlynarcikova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava, 84505, Slovakia.
| | - Sona Scsukova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava, 84505, Slovakia
| |
Collapse
|
43
|
Suryan V, Chandra NC. Cholesterol and Cytokines: Molecular Links to Atherosclerosis and Carcinogenesis. Cell Biochem Biophys 2024; 82:1837-1844. [PMID: 38943010 DOI: 10.1007/s12013-024-01383-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2024] [Indexed: 06/30/2024]
Abstract
An increase of cholesterol concentration within the artery obstructs arterial blood flow once it deposits alongside the arterial wall. This results in atherosclerosis. Carcinogenesis causes a quicker clearance of vascular cholesterol to meet the demands of tumour cell development. Both illnesses have an increased concentration of pro-inflammatory cytokines in the blood. To search the comparative characteristics of cholesterol and pro-inflammatory cytokines in the pathogenesis of atherosclerosis and carcinogenesis, a comprehensive online survey using MEDLINE, Scopus, PubMed, and Google Scholar was conducted for relevant journals with key search term cholesterol and cytokines in atherosclerotic and cancerous patients. According to reports, hypercholesterolaemia related dyslipidemia causes atherosclerosis in blood arteries and hypercholesterolaemia in cell nucleus is a reason for developing carcinogenesis. It is also noted that pro-inflammatory cytokines are involved in both of the aforementioned pathogenesis. Changes in anti-inflammatory cytokines are only the characteristic features of each kind. Thus, Cholesterol and pro-inflammatory cytokines are intensely interlinked in the genesis of atherosclerotic and carcinogenic consequences.
Collapse
Affiliation(s)
- Varsha Suryan
- Department of Biochemistry, Faculty of Medicine & Health Sciences, Shree Guru Gobind Singh Tricentenary University, Budhera, Gurugram (Delhi-NCR), Haryana, 122505, India
- Department of Paramedical Science, Faculty of Allied Health Sciences, Shree Guru Gobind Singh Tricentenary University, Budhera, Gurugram (Delhi-NCR), Haryana, 122505, India
| | - Nimai Chand Chandra
- Department of Biochemistry, Faculty of Medicine & Health Sciences, Shree Guru Gobind Singh Tricentenary University, Budhera, Gurugram (Delhi-NCR), Haryana, 122505, India.
| |
Collapse
|
44
|
Bhol NK, Bhanjadeo MM, Singh AK, Dash UC, Ojha RR, Majhi S, Duttaroy AK, Jena AB. The interplay between cytokines, inflammation, and antioxidants: mechanistic insights and therapeutic potentials of various antioxidants and anti-cytokine compounds. Biomed Pharmacother 2024; 178:117177. [PMID: 39053423 DOI: 10.1016/j.biopha.2024.117177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/03/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024] Open
Abstract
Cytokines regulate immune responses essential for maintaining immune homeostasis, as deregulated cytokine signaling can lead to detrimental outcomes, including inflammatory disorders. The antioxidants emerge as promising therapeutic agents because they mitigate oxidative stress and modulate inflammatory pathways. Antioxidants can potentially ameliorate inflammation-related disorders by counteracting excessive cytokine-mediated inflammatory responses. A comprehensive understanding of cytokine-mediated inflammatory pathways and the interplay with antioxidants is paramount for developing natural therapeutic agents targeting inflammation-related disorders and helping to improve clinical outcomes and enhance the quality of life for patients. Among these antioxidants, curcumin, vitamin C, vitamin D, propolis, allicin, and cinnamaldehyde have garnered attention for their anti-inflammatory properties and potential therapeutic benefits. This review highlights the interrelationship between cytokines-mediated disorders in various diseases and therapeutic approaches involving antioxidants.
Collapse
Affiliation(s)
- Nitish Kumar Bhol
- Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar, Odisha 751004, India
| | | | - Anup Kumar Singh
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India
| | - Umesh Chandra Dash
- Environmental Biotechnology Laboratory, KIIT School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Odisha, India
| | - Rakesh Ranjan Ojha
- Department of Bioinformatics, BJB (A) College, Bhubaneswar, Odisha-751014, India
| | - Sanatan Majhi
- Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar, Odisha 751004, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Medical Sciences, Faculty of Medicine, University of Oslo, Norway.
| | - Atala Bihari Jena
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India.
| |
Collapse
|
45
|
Obara H, Tatara Y, Monzen S, Murakami S, Yamamoto H, Kimura N, Suzuki M, Komai F, Narita M, Hatayama Y, Aoki M. Exploring predictive molecules of acute adverse events in response to volumetric‑modulated arc therapy for prostate cancer using urinary metabolites. Mol Clin Oncol 2024; 21:62. [PMID: 39071973 PMCID: PMC11273245 DOI: 10.3892/mco.2024.2760] [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: 02/28/2024] [Accepted: 06/17/2024] [Indexed: 07/30/2024] Open
Abstract
Volumetric-modulated arc therapy (VMAT) is a radiotherapy technique used to treat patients with localized prostate cancer, which is frequently associated with acute adverse events (AEs) that can affect subsequent treatment. Notably, the radiation dose of VMAT can be tailored to each patient. In the present study, a retrospective analysis was performed to predict acute AEs in response to a therapeutic high radiation dose rate based on urinary metabolomic molecules, which are easily collected as noninvasive biosamples. Urine samples from 11 patients with prostate cancer who were treated with VMAT (76 Gy/38 fractions) were collected. The study found that seven patients (~64%) exhibited genitourinary toxicity (Grade 1) and four patients had no AEs. A total of 630 urinary metabolites were then analyzed using a mass spectrometer (QTRAP6500+; AB SCIEX), and 234 relevant molecules for biological and clinical applications were extracted from the absolute quantified metabolite values using the MetaboINDICATOR tool. In the Grade 1 acute AE group, there was a significant negative correlation (rs=-0.297, P<0.05) between the number of VMAT fractions and total phospholipase A2 activity in the urine. Additionally, patients with Grade 1 AEs exhibited a decrease in PC aa C40:1, a phospholipid. These findings suggested that specific lipids found in urinary metabolites may serve as predictive biomarkers for acute AEs in response to external radiotherapy.
Collapse
Affiliation(s)
- Hideki Obara
- Department of Radiology, Hirosaki University Hospital, Hirosaki, Aomori 036-8563, Japan
| | - Yota Tatara
- Department of Stress Response Science, Biomedical Research Center, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Satoru Monzen
- Department of Radiation Science, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036-8564, Japan
- Research Center for Biomedical Sciences, Hirosaki University, Hirosaki, Aomori 036-8564, Japan
| | - Sho Murakami
- Department of Radiology, Hirosaki University Hospital, Hirosaki, Aomori 036-8563, Japan
| | - Hiroki Yamamoto
- Department of Radiology, Hirosaki University Hospital, Hirosaki, Aomori 036-8563, Japan
| | - Naoki Kimura
- Department of Radiology, Hirosaki University Hospital, Hirosaki, Aomori 036-8563, Japan
| | - Masashi Suzuki
- Department of Radiology, Hirosaki University Hospital, Hirosaki, Aomori 036-8563, Japan
| | - Fumio Komai
- Department of Radiology, Hirosaki University Hospital, Hirosaki, Aomori 036-8563, Japan
| | - Masataka Narita
- Department of Radiology, Hirosaki University Hospital, Hirosaki, Aomori 036-8563, Japan
| | - Yoshiomi Hatayama
- Department of Radiology, Hirosaki University Hospital, Hirosaki, Aomori 036-8563, Japan
| | - Masahiko Aoki
- Department of Radiology, Hirosaki University Hospital, Hirosaki, Aomori 036-8563, Japan
| |
Collapse
|
46
|
Maeshima Y, Kataoka TR, Vandenbon A, Hirata M, Takeuchi Y, Suzuki Y, Fukui Y, Kawashima M, Takada M, Ibi Y, Haga H, Morita S, Toi M, Kawaoka S, Kawaguchi K. Intra-patient spatial comparison of non-metastatic and metastatic lymph nodes reveals the reduction of CD169 + macrophages by metastatic breast cancers. EBioMedicine 2024; 107:105271. [PMID: 39173531 PMCID: PMC11382037 DOI: 10.1016/j.ebiom.2024.105271] [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: 12/20/2023] [Revised: 06/06/2024] [Accepted: 07/25/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Breast cancer cells suppress the host immune system to efficiently invade the lymph nodes; however, the underlying mechanism remains incompletely understood. Here, we aimed to comprehensively characterise the effects of breast cancers on immune cells in the lymph nodes. METHODS We collected non-metastatic and metastatic lymph node samples from 6 patients with breast cancer with lymph node metastasis. We performed bulk transcriptomics, spatial transcriptomics, and imaging mass cytometry to analyse the obtained lymph nodes. Furthermore, we conducted histological analyses against a larger patient cohort (474 slices from 58 patients). FINDINGS The comparison between paired lymph nodes with and without metastasis from the same patients demonstrated that the number of CD169+ lymph node sinus macrophages, an initiator of anti-cancer immunity, was reduced in metastatic lymph nodes (36.7 ± 21.1 vs 7.3 ± 7.0 cells/mm2, p = 0.0087), whereas the numbers of other major immune cell types were unaltered. We also detected that the infiltration of CD169+ macrophages into metastasised cancer tissues differed by section location within tumours, suggesting that CD169+ macrophages were gradually decreased after anti-cancer reactions. Furthermore, CD169+ macrophage elimination was prevalent in major breast cancer subtypes and correlated with breast cancer staging (p = 0.022). INTERPRETATION We concluded that lymph nodes with breast cancer metastases have fewer CD169+ macrophages, which may be detrimental to the activity of anti-cancer immunity. FUNDING JSPS KAKENHI (16H06279, 20H03451, 20H04842, 22H04925, 19K16770, and 21K15530, 24K02236), JSPS Fellows (JP22KJ1822), AMED (JP21ck0106698), JST FOREST (JPMJFR2062), Caravel, Co., Ltd, Japan Foundation for Applied Enzymology, and Sumitomo Pharma Co., Ltd. under SKIPS.
Collapse
Affiliation(s)
- Yurina Maeshima
- Department of Breast Surgery, Kyoto University Hospital, Graduate School of Medicine, Shogoin Sakyo-ku, Kyoto 606-8507, Japan; Inter-Organ Communication Research Team, Institute for Frontier Life and Medical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tatsuki R Kataoka
- Department of Pathology, Iwate Medical University, Yahaba-cho, Shiwa-gun, Iwate Prefecture 028-3694, Japan
| | - Alexis Vandenbon
- Laboratory of Tissue Homeostasis, Institute for Life and Medical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan; Institute for Liberal Arts and Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Masahiro Hirata
- Department of Diagnostic Pathology, Kyoto University, Shogoin Sakyo-ku, Kyoto 606-8507, Japan
| | - Yasuhide Takeuchi
- Department of Diagnostic Pathology, Kyoto University, Shogoin Sakyo-ku, Kyoto 606-8507, Japan
| | - Yutaka Suzuki
- Graduate School of Frontier Science, The University of Tokyo, Chiba 277-8562, Japan
| | - Yukiko Fukui
- Department of Breast Surgery, Kyoto University Hospital, Graduate School of Medicine, Shogoin Sakyo-ku, Kyoto 606-8507, Japan
| | - Masahiro Kawashima
- Department of Breast Surgery, Kyoto University Hospital, Graduate School of Medicine, Shogoin Sakyo-ku, Kyoto 606-8507, Japan
| | - Masahiro Takada
- Department of Breast Surgery, Kyoto University Hospital, Graduate School of Medicine, Shogoin Sakyo-ku, Kyoto 606-8507, Japan
| | - Yumiko Ibi
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Shogoin Sakyo-ku, Kyoto 606-8507, Japan
| | - Hironori Haga
- Department of Diagnostic Pathology, Kyoto University, Shogoin Sakyo-ku, Kyoto 606-8507, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Shogoin Sakyo-ku, Kyoto 606-8507, Japan
| | - Masakazu Toi
- Department of Breast Surgery, Kyoto University Hospital, Graduate School of Medicine, Shogoin Sakyo-ku, Kyoto 606-8507, Japan; Tokyo Metropolitan Cancer and Infectious Disease Center, Komagome Hospital, Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan
| | - Shinpei Kawaoka
- Inter-Organ Communication Research Team, Institute for Frontier Life and Medical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan; Department of Integrative Bioanalytics, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan.
| | - Kosuke Kawaguchi
- Department of Breast Surgery, Kyoto University Hospital, Graduate School of Medicine, Shogoin Sakyo-ku, Kyoto 606-8507, Japan; Department of Breast Surgery, Breast Center, Mie University, Mie 514-0102, Japan.
| |
Collapse
|
47
|
Prodi E, Neri D, De Luca R. Tumor-Homing Antibody-Cytokine Fusions for Cancer Therapy. Onco Targets Ther 2024; 17:697-715. [PMID: 39224695 PMCID: PMC11368152 DOI: 10.2147/ott.s480787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
Recombinant cytokine products have emerged as a promising avenue in cancer therapy due to their capacity to modulate and enhance the immune response against tumors. However, their clinical application is significantly hindered by systemic toxicities already at low doses, thus preventing escalation to therapeutically active regimens. One promising approach to overcoming these limitations is using antibody-cytokine fusion proteins (also called immunocytokines). These biopharmaceuticals leverage the targeting specificity of antibodies to deliver cytokines directly to the tumor microenvironment, thereby reducing systemic exposure and enhancing the therapeutic index. This review comprehensively examines the development and potential of antibody-cytokine fusion proteins in cancer therapy. It explores the molecular characteristics that influence the performance of these fusion proteins, and it highlights key findings from preclinical and clinical studies, illustrating the potential of immunocytokines to improve treatment outcomes in cancer patients. Recent advancements in the field, such as novel engineering strategies and combination strategies to enhance the efficacy and safety of immunocytokines, are also discussed. These innovations offer new opportunities to optimize this class of biotherapeutics, making them a more viable and effective option for cancer treatment. As the field continues to evolve, understanding the critical factors that influence the performance of immunocytokines will be essential for successfully translating these therapies into clinical practice.
Collapse
Affiliation(s)
- Eleonora Prodi
- Philochem AG, Otelfingen, 8112, Switzerland
- University of Trento, Italy, CiBIO (Department of Cellular, Computational and Integrative Biology), Povo, 38123, Trento
| | - Dario Neri
- Philogen Spa, Siena, 53100, Italy
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | | |
Collapse
|
48
|
Sakellakis M, Reet J, Kladas M, Hoge G, Chalkias A, Radulovic M. Cancer-Induced Resting Sinus Tachycardia: An Overlooked Clinical Diagnosis. Oncol Rev 2024; 18:1439415. [PMID: 39156014 PMCID: PMC11327047 DOI: 10.3389/or.2024.1439415] [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: 05/27/2024] [Accepted: 07/23/2024] [Indexed: 08/20/2024] Open
Abstract
Elevated resting heart rate is frequently observed in cancer patients, and is associated with increased mortality. Although specific chemotherapeutic agents can induce cardiotoxicity, the presence of sinus tachycardia in chemotherapy-naive patients suggests other factors likely contribute to this clinical presentation. Despite its prevalence, cancer-associated resting sinus tachycardia has not been fully recognized and comprehensively described as a separate clinical entity. Secondary effects of cancer, especially structural cardiac changes, secretory factors (inflammatory cytokines), and thromboembolic disease can cause resting tachycardia. Alternatively, rapid heart rate may reflect compensatory mechanisms responding to increased metabolic demands, raised cardiac output states, and even pain. Hence, cancer-associated tachycardia presents a clinical dilemma; acute life-threatening conditions (such as sepsis, pulmonary embolism, etc.) must be ruled out, but cancer itself can explain resting sinus tachycardia and more conservative management can avoid unnecessary testing, cost and patient stress. Furthermore, identification and management of cardiac conditions associated with cancer may improve survival and the quality of life of cancer patients.
Collapse
Affiliation(s)
- Minas Sakellakis
- Department of Medicine, Jacobi Medical Center/North Central Bronx Hospital, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Jashan Reet
- Department of Medicine, Jacobi Medical Center/North Central Bronx Hospital, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Michail Kladas
- Department of Medicine, Jacobi Medical Center/North Central Bronx Hospital, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Gregory Hoge
- Department of Medicine, Jacobi Medical Center/North Central Bronx Hospital, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Athanasios Chalkias
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Outcomes Research Consortium, Cleveland, OH, United States
| | - Miroslav Radulovic
- Department of Medicine, Jacobi Medical Center/North Central Bronx Hospital, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
49
|
Garrone O, Paccagnella M, Abbona A, Ruatta F, Vanella P, Denaro N, Tomasello G, Croce N, Barbin F, Rossino MG, La Porta CAM, Sapino A, Torri V, Albini A, Merlano MC. Moderate physical activity during neoadjuvant chemotherapy in breast cancer patients: effect on cancer-related inflammation and pathological complete response-the Neo-Runner study. ESMO Open 2024; 9:103665. [PMID: 39121813 PMCID: PMC11364046 DOI: 10.1016/j.esmoop.2024.103665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 07/06/2024] [Accepted: 07/15/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Physical activity (PA) reduces the risk of developing breast cancer (BC) and mortality rate in BC patients starting PA after diagnosis. Immunomodulation is considered responsible for these effects. However, limited data exist on the immunomodulation induced by moderate PA (mPA) during neoadjuvant chemotherapy (NACT). We have investigated the longitudinal change of cytokines during NACT alone or combined with mPA. MATERIALS AND METHODS Twenty-three cytokines were analyzed in BC patients at consecutive timepoints: at baseline (T0), before starting mPA (T1), before surgery (T2), and after surgery (T3). mPA consisted of 3-weekly brisk-walking sessions for 9-10 consecutive weeks. RESULTS Ninety-two patients were assessed: 21 patients refused mPA (untrained) and 71 agreed (trained). At T1, NACT induced significant up-regulation of interleukin (IL)-5, IL-6, IL-15, chemokine ligand (CCL)-2, interferon-γ, and C-X-C motif ligand (CXCL)-10 and reduction of expression of IL-13 and CCL-22. At T2, NACT and mPA induced up-regulation of IL-21, CCL-2, and tumor necrosis factor-α and reduction of expression of IL-8, IL-15, vascular endothelial growth factor, and soluble interleukin 6 receptor. Only CXCL-10 increased in untrained patients. A cytokine score (CS) was created to analyze, all together, the changes between T1 and T2. At T2 the CS decreased in trained and increased in untrained patients. We clustered the patients using cytokines and predictive factors and identified two clusters. The cluster A, encompassing 90% of trained patients, showed more pathological complete response (pCR) compared to the cluster B: 78% versus 22%, respectively. CONCLUSIONS mPA interacts with NACT inducing CS reduction in trained patients not observed in untrained patients, suggesting a reduction of inflammation, notwithstanding chemotherapy. This effect may contribute to the higher rate of pCR observed in the cluster A, including most trained patients.
Collapse
Affiliation(s)
- O Garrone
- Department of Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan.
| | - M Paccagnella
- Translational Oncology ARCO Foundation, Cuneo. https://twitter.com/matteo_babeuf
| | - A Abbona
- Translational Oncology ARCO Foundation, Cuneo. https://twitter.com/AbbonaAndr36863
| | - F Ruatta
- Department of Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan. https://twitter.com/fiorella_ruatta
| | - P Vanella
- Department of Medical Oncology, S. Croce e Carle Teaching Hospital, Cuneo
| | - N Denaro
- Department of Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan
| | - G Tomasello
- Medical Oncology, ASST Ospedale Maggiore Crema, Crema. https://twitter.com/glucatom
| | - N Croce
- Department of Medical Oncology, S. Croce e Carle Teaching Hospital, Cuneo
| | - F Barbin
- Department of Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan
| | - M G Rossino
- Department of Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan
| | - C A M La Porta
- Center for Complexity and Biosystems, University of Milan, Milan; Department of Environmental Science and Policy, University of Milan, Milan. https://twitter.com/CAMLaPorta
| | - A Sapino
- Scientific Direction, Candiolo Cancer Institute, FPO-IRCCS Candiolo, Turin; Department of Medical Science, University of Turin, Turin. https://twitter.com/sapino58
| | - V Torri
- Mario Negri Institute for Pharmacological Research, Milan. https://twitter.com/ValterTorri
| | - A Albini
- European Institute of Oncology, Milan, Italy. https://twitter.com/adrianaalbini1
| | - M C Merlano
- Scientific Direction, Candiolo Cancer Institute, FPO-IRCCS Candiolo, Turin
| |
Collapse
|
50
|
Dawalibi A, Alosaimi AA, Mohammad KS. Balancing the Scales: The Dual Role of Interleukins in Bone Metastatic Microenvironments. Int J Mol Sci 2024; 25:8163. [PMID: 39125732 PMCID: PMC11311339 DOI: 10.3390/ijms25158163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
Bone metastases, a common and debilitating consequence of advanced cancers, involve a complex interplay between malignant cells and the bone microenvironment. Central to this interaction are interleukins (ILs), a group of cytokines with critical roles in immune modulation and inflammation. This review explores the dualistic nature of pro-inflammatory and anti-inflammatory interleukins in bone metastases, emphasizing their molecular mechanisms, pathological impacts, and therapeutic potential. Pro-inflammatory interleukins, such as IL-1, IL-6, and IL-8, have been identified as key drivers in promoting osteoclastogenesis, tumor proliferation, and angiogenesis. These cytokines create a favorable environment for cancer cell survival and bone degradation, contributing to the progression of metastatic lesions. Conversely, anti-inflammatory interleukins, including IL-4, IL-10, and IL-13, exhibit protective roles by modulating immune responses and inhibiting osteoclast activity. Understanding these opposing effects is crucial for developing targeted therapies aimed at disrupting the pathological processes in bone metastases. Key signaling pathways, including NF-κB, JAK/STAT, and MAPK, mediate the actions of these interleukins, influencing tumor cell survival, immune cell recruitment, and bone remodeling. Targeting these pathways presents promising therapeutic avenues. Current treatment strategies, such as the use of denosumab, tocilizumab, and emerging agents like bimekizumab and ANV419, highlight the potential of interleukin-targeted therapies in mitigating bone metastases. However, challenges such as therapeutic resistance, side effects, and long-term efficacy remain significant hurdles. This review also addresses the potential of interleukins as diagnostic and prognostic biomarkers, offering insights into patient stratification and personalized treatment approaches. Interleukins have multifaceted roles that depend on the context, including the environment, cell types, and cellular interactions. Despite substantial progress, gaps in research persist, particularly regarding the precise mechanisms by which interleukins influence the bone metastatic niche and their broader clinical implications. While not exhaustive, this overview underscores the critical roles of interleukins in bone metastases and highlights the need for continued research to fully elucidate their complex interactions and therapeutic potential. Addressing these gaps will be essential for advancing our understanding and treatment of bone metastases in cancer patients.
Collapse
Affiliation(s)
- Ahmad Dawalibi
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| | - Amal Ahmed Alosaimi
- College of Medicine, Imam Mohammad Ibn Saud Islamic University, Riyadh 11432, Saudi Arabia;
| | - Khalid S. Mohammad
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| |
Collapse
|