1
|
Prkačin I, Mokos M, Ferara N, Šitum M. Melanoma's New Frontier: Exploring the Latest Advances in Blood-Based Biomarkers for Melanoma. Cancers (Basel) 2024; 16:4219. [PMID: 39766118 PMCID: PMC11727356 DOI: 10.3390/cancers16244219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/06/2025] Open
Abstract
Melanoma is one of the most malignant cancers, and the global incidence of cutaneous melanoma is increasing. While melanomas are highly prone to metastasize if diagnosed late, early detection and treatment significantly reduce the risk of mortality. Identifying patients at higher risk of metastasis, who might benefit from early adjuvant therapies, is particularly important, especially with the advent of new melanoma treatments. Therefore, there is a pressing need to develop additional prognostic biomarkers for melanoma to improve early stratification of patients and accurately identify high-risk subgroups, ultimately enabling more effective personalized treatments. Recent advances in melanoma therapy, including targeted treatments and immunotherapy, have underscored the importance of biomarkers in determining prognosis and predicting treatment response. The clinical application of these markers holds the potential for significant advancements in melanoma management. Various tumor-derived genetic, proteomic, and cellular components are continuously released into the bloodstream of cancer patients. These molecules, including circulating tumor DNA and RNA, proteins, tumor cells, and immune cells, are emerging as practical and precise liquid biomarkers for cancer. In the current era of effective molecular-targeted therapies and immunotherapies, there is an urgent need to integrate these circulating biomarkers into clinical practice to facilitate personalized treatment. This review highlights recent discoveries in circulating melanoma biomarkers, explores the challenges and potentials of emerging technologies for liquid biomarker discovery, and discusses future directions in melanoma biomarker research.
Collapse
Affiliation(s)
- Ivana Prkačin
- Department of Dermatology and Venereology, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia; (M.M.); (N.F.); (M.Š.)
- School of Medicine, University of Split, 21000 Split, Croatia
| | - Mislav Mokos
- Department of Dermatology and Venereology, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia; (M.M.); (N.F.); (M.Š.)
| | - Nikola Ferara
- Department of Dermatology and Venereology, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia; (M.M.); (N.F.); (M.Š.)
| | - Mirna Šitum
- Department of Dermatology and Venereology, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia; (M.M.); (N.F.); (M.Š.)
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Croatian Academy of Sciences and Arts, 10000 Zagreb, Croatia
| |
Collapse
|
2
|
Sosa Cuevas E, Mouret S, Vayssière G, Kerboua S, Girard P, Molens JP, Manceau M, Charles J, Saas P, Aspord C. Circulating immune landscape in melanoma patients undergoing anti-PD1 therapy reveals key immune features according to clinical response to treatment. Front Immunol 2024; 15:1507938. [PMID: 39687620 PMCID: PMC11646980 DOI: 10.3389/fimmu.2024.1507938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Introduction Immune checkpoint blockers (ICB) bring unprecedented clinical success, yet many patients endure immune mediated adverse effects and/or fail to respond. Predictive signatures of response to ICB and mechanisms of clinical efficacy or failure remain understudied. DC subsets, in network with conventional αβ T (Tconv), NK, γδ T and iNKT cells, harbor pivotal roles in tumor control, yet their involvement in response to ICB remained underexplored. Methods We performed an extensive longitudinal monitoring of circulating immune cells from melanoma patients treated with first-line anti-PD1, before (T0) and during treatment. We assessed the phenotypic and functional features of DC and effector cells' subsets by multi-parametric flow cytometry and ProcartaPlex® dosages. Results We revealed differences according to response to treatment and modulations of patterns during treatment, highlighting a strong link between the immune landscape and the outcome of anti-PD1 therapy. Responders exhibited higher frequencies of circulating cDC1s, CD8+ T cells, and γδ2+ T cells in central memory (CM) stage. Notably, we observed a distinct remodeling of ICP expression profile, activation status and natural cytotoxicity receptor patterns of immune subsets during treatment. Anti-PD1 modulated DCs' functionality and triggered deep changes in the functional orientation of Tconv and γδT cells. Discussion Overall, our work provides new insights into the immunological landscape sustaining favorable clinical responses or resistance to first-line anti-PD1 therapy in melanoma patients. Such exploration participates in uncovering the mechanism of action of anti-PD1, discovering innovative predictive signatures of response, and paves the way to design pertinent combination strategies to improve patient clinical benefits in the future.
Collapse
Affiliation(s)
- Eleonora Sosa Cuevas
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Stéphane Mouret
- Dermatology, Allergology & Photobiology Department, CHU Grenoble Alpes, Univ. Grenoble Alpes, Grenoble, France
| | - Guillaume Vayssière
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Siham Kerboua
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Pauline Girard
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Jean-Paul Molens
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Marc Manceau
- Department of Medicine, Clinical Investigation Center, CHU Grenoble Alpes, Univ. Grenoble Alpes, Grenoble, France
| | - Julie Charles
- Dermatology, Allergology & Photobiology Department, CHU Grenoble Alpes, Univ. Grenoble Alpes, Grenoble, France
| | - Philippe Saas
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Caroline Aspord
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| |
Collapse
|
3
|
Gorgulho J, Loosen SH, Masood R, Giehren F, Pagani F, Buescher G, Kocheise L, Joerg V, Schmidt C, Schulze K, Roderburg C, Kinkel E, Fritzsche B, Wehmeyer S, Schmidt B, Kachel P, Rolling C, Götze J, Busch A, Sinn M, Pereira-Veiga T, Wikman H, Geffken M, Peine S, Matschl U, Altfeld M, Huber S, Lohse AW, Beier F, Brümmendorf TH, Bokemeyer C, Luedde T, von Felden J. Soluble and EV-bound CD27 act as antagonistic biomarkers in patients with solid tumors undergoing immunotherapy. J Exp Clin Cancer Res 2024; 43:298. [PMID: 39511626 PMCID: PMC11545160 DOI: 10.1186/s13046-024-03215-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND The major breakthrough in cancer therapy with immune checkpoint inhibitors (ICIs) has highlighted the important role of immune checkpoints in antitumoral immunity. However, most patients do not achieve durable responses, making biomarker research in this setting essential. CD27 is a well known costimulatory molecule, however the impact of its soluble form in ICI is poorly investigated. Therefore, we aimed at testing circulating concentrations of soluble CD27 (sCD27) and CD27 bound to extracellular vesicles (EVs) as potential biomarkers to predict response and overall survival (OS) in patients undergoing ICI. METHODS Serum and plasma levels of sCD27 were assessed by immunoassay in three patient cohorts (n = 187) with advanced solid malignancies including longitudinal samples (n = 126): a training (n = 84, 210 specimens, Aachen ICI) and validation cohort (n = 70, 70 specimens, Hamburg ICI), both treated with ICI therapy, and a second independent validation cohort (n = 33, 33 specimens, Hamburg non-ICI) undergoing systemic therapy without any ICI. In a subset (n = 36, 36 baseline and 108 longitudinal specimens), EV-bound CD27 from serum was measured, while EV characterization studies were conducted on a fourth cohort (n = 45). RESULTS In the Aachen and Hamburg ICI cohorts, patients with lower circulating sCD27 levels before and during ICI therapy had a significantly longer progression-free survival (PFS) and OS compared to patients with higher levels, a finding that was confirmed by multivariate analysis (MVA) (Aachen ICI: pPFS = 0.012, pOS = 0.001; Hamburg ICI: pPFS = 0.040, pOS = 0.004) and after randomly splitting both cohorts into training and validation. This phenomenon was not observed in the Hamburg non-ICI cohort, providing a rationale for the predictive biomarker role of sCD27 in immune checkpoint blockade. Remarkably, EV-bound CD27 baseline levels and dynamics during ICI therapy also emerged as potent predictive biomarkers, acting however antagonistically to soluble sCD27, i.e. higher levels were associated with PFS and OS benefit. Combining both molecules ("multi-CD27" score) enhanced the predictive ability (HRPFS: 17.21 with p < 0.001, HROS: 6.47 with p = 0.011). CONCLUSION Soluble and EV-bound CD27 appear to have opposing immunomodulatory functions and may represent easily measurable, non-invasive prognostic markers to predict response and survival in patients undergoing ICI therapy.
Collapse
Affiliation(s)
- Joao Gorgulho
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- University Cancer Center Hamburg - Hubertus Wald Tumorzentrum, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Sven H Loosen
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Center for Integrated Oncology, Aachen-Bonn-Cologne-Düsseldorf (CIOABCD), Aachen, Germany
| | - Ramsha Masood
- I. Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Franziska Giehren
- I. Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Francesca Pagani
- I. Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Gustav Buescher
- I. Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Lorenz Kocheise
- I. Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Vincent Joerg
- I. Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Constantin Schmidt
- I. Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Kornelius Schulze
- I. Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany
| | - Christoph Roderburg
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Center for Integrated Oncology, Aachen-Bonn-Cologne-Düsseldorf (CIOABCD), Aachen, Germany
| | - Eva Kinkel
- University Cancer Center Hamburg - Hubertus Wald Tumorzentrum, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Britta Fritzsche
- University Cancer Center Hamburg - Hubertus Wald Tumorzentrum, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Simon Wehmeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Benjamin Schmidt
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Paul Kachel
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Christina Rolling
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Julian Götze
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Alina Busch
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Marianne Sinn
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Thais Pereira-Veiga
- Department of Tumor Biology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Harriet Wikman
- Department of Tumor Biology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Maria Geffken
- Institute of Transfusion Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sven Peine
- Institute of Transfusion Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | - Samuel Huber
- I. Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany
| | - Ansgar W Lohse
- I. Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany
| | - Fabian Beier
- Center for Integrated Oncology, Aachen-Bonn-Cologne-Düsseldorf (CIOABCD), Aachen, Germany
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Tim H Brümmendorf
- Center for Integrated Oncology, Aachen-Bonn-Cologne-Düsseldorf (CIOABCD), Aachen, Germany
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- University Cancer Center Hamburg - Hubertus Wald Tumorzentrum, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- Center for Integrated Oncology, Aachen-Bonn-Cologne-Düsseldorf (CIOABCD), Aachen, Germany.
| | - Johann von Felden
- I. Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany.
| |
Collapse
|
4
|
Arghidash F, Javid-Naderi MJ, Gheybi F, Gholamhosseinian H, Kesharwani P, Sahebkar A. Exploring the multifaceted effects of silymarin on melanoma: Focusing on the role of lipid-based nanocarriers. J Drug Deliv Sci Technol 2024; 99:105950. [DOI: 10.1016/j.jddst.2024.105950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
5
|
Zhao Z, Huo Y, Du Y, Huang Y, Liu H, Zhang C, Yan J. A neutrophil extracellular trap-related risk score predicts prognosis and characterizes the tumor microenvironment in multiple myeloma. Sci Rep 2024; 14:2264. [PMID: 38278930 PMCID: PMC10817968 DOI: 10.1038/s41598-024-52922-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/25/2024] [Indexed: 01/28/2024] Open
Abstract
Multiple myeloma (MM) is a distinguished hematologic malignancy, with existing studies elucidating its interaction with neutrophil extracellular traps (NETs), which may potentially facilitate tumor growth. However, systematic investigations into the role of NETs in MM remain limited. Utilizing the single-cell dataset GSE223060, we discerned active NET cell subgroups, namely neutrophils, monocytes, and macrophages. A transcriptional trajectory was subsequently constructed to comprehend the progression of MM. Following this, an analysis of cellular communication in MM was conducted with a particular emphasis on neutrophils, revealing an augmentation in interactions albeit with diminished strength, alongside abnormal communication links between neutrophils and NK cells within MM samples. Through the intersection of differentially expressed genes (DEGs) between NET active/inactive cells and MM versus healthy samples, a total of 316 genes were identified. This led to the development of a 13-gene risk model for prognostic prediction based on overall survival, utilizing transcriptomics dataset GSE136337. The high-risk group manifested altered immune infiltration and heightened sensitivity to chemotherapy. A constructed nomogram for predicting survival probabilities demonstrated encouraging AUCs for 1, 3, and 5-year survival predictions. Collectively, our findings unveil a novel NET-related prognostic signature for MM, thereby providing a potential avenue for therapeutic exploration.
Collapse
Affiliation(s)
- Zhijia Zhao
- Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Yuan Huo
- Diamond Bay Institute of Hematology, The Second Hospital of Dalian Medical University, Dalian, 116031, China
| | - Yufeng Du
- Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, The Second Hospital of Dalian Medical University, Dalian, 116023, China
- Blood Stem Cell Transplantation Institute of Dalian Medical University, Dalian, 116023, China
| | - Yanan Huang
- Diamond Bay Institute of Hematology, The Second Hospital of Dalian Medical University, Dalian, 116031, China
| | - Hongchen Liu
- Diamond Bay Institute of Hematology, The Second Hospital of Dalian Medical University, Dalian, 116031, China
| | - Chengtao Zhang
- Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, The Second Hospital of Dalian Medical University, Dalian, 116023, China.
- Blood Stem Cell Transplantation Institute of Dalian Medical University, Dalian, 116023, China.
| | - Jinsong Yan
- Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, The Second Hospital of Dalian Medical University, Dalian, 116023, China.
- Diamond Bay Institute of Hematology, The Second Hospital of Dalian Medical University, Dalian, 116031, China.
- Blood Stem Cell Transplantation Institute of Dalian Medical University, Dalian, 116023, China.
- Pediatric Oncology and Hematology Center, The Second Hospital of Dalian Medical University, Dalian, 116023, China.
| |
Collapse
|
6
|
Caraban BM, Aschie M, Deacu M, Cozaru GC, Pundiche MB, Orasanu CI, Voda RI. A Narrative Review of Current Knowledge on Cutaneous Melanoma. Clin Pract 2024; 14:214-241. [PMID: 38391404 PMCID: PMC10888040 DOI: 10.3390/clinpract14010018] [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: 12/20/2023] [Revised: 01/21/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Cutaneous melanoma is a public health problem. Efforts to reduce its incidence have failed, as it continues to increase. In recent years, many risk factors have been identified. Numerous diagnostic systems exist that greatly assist in early clinical diagnosis. The histopathological aspect illustrates the grim nature of these cancers. Currently, pathogenic pathways and the tumor microclimate are key to the development of therapeutic methods. Revolutionary therapies like targeted therapy and immune checkpoint inhibitors are starting to replace traditional therapeutic methods. Targeted therapy aims at a specific molecule in the pathogenic chain to block it, stopping cell growth and dissemination. The main function of immune checkpoint inhibitors is to boost cellular immunity in order to combat cancer cells. Unfortunately, these therapies have different rates of effectiveness and side effects, and cannot be applied to all patients. These shortcomings are the basis of increased incidence and mortality rates. This study covers all stages of the evolutionary sequence of melanoma. With all these data in front of us, we see the need for new research efforts directed at therapies that will bring greater benefits in terms of patient survival and prognosis, with fewer adverse effects.
Collapse
Affiliation(s)
- Bogdan Marian Caraban
- Clinical Department of Plastic Surgery, Microsurgery-Reconstructive, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
- Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
| | - Mariana Aschie
- Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
- Clinical Service of Pathology, Departments of Pathology, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
- Academy of Medical Sciences of Romania, 030171 Bucharest, Romania
- The Romanian Academy of Scientists, 030167 Bucharest, Romania
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), "Ovidius" University of Constanta, 900591 Constanta, Romania
| | - Mariana Deacu
- Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
- Clinical Service of Pathology, Departments of Pathology, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
| | - Georgeta Camelia Cozaru
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), "Ovidius" University of Constanta, 900591 Constanta, Romania
- Clinical Service of Pathology, Departments of Genetics, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
| | - Mihaela Butcaru Pundiche
- Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
- Clinical Department of General Surgery, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
| | - Cristian Ionut Orasanu
- Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
- Clinical Service of Pathology, Departments of Pathology, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), "Ovidius" University of Constanta, 900591 Constanta, Romania
| | - Raluca Ioana Voda
- Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
- Clinical Service of Pathology, Departments of Pathology, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), "Ovidius" University of Constanta, 900591 Constanta, Romania
| |
Collapse
|
7
|
Guo W, Wang X, Zhang Y, Liu H, Ma S, Guan F. Construction and validation of a novel prognostic signature for cutaneous melanoma based on ferroptosis-related genes. Heliyon 2023; 9:e15725. [PMID: 37159694 PMCID: PMC10163649 DOI: 10.1016/j.heliyon.2023.e15725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/09/2023] [Accepted: 04/19/2023] [Indexed: 05/11/2023] Open
Abstract
Ferroptosis, a recently uncovered iron-dependent, non-apoptotic cell death process, has been increasingly linked to cancer development. In this study, our objective was to develop a prognostic model centered on ferroptosis-related genes (FRGs) and assess its efficacy as an overall survival (OS) prediction biomarker. We conducted a systematic analysis of cutaneous melanoma (CM) and devised a novel ferroptosis-related prognostic signature (FRGSig) using the TCGA database. An independent dataset from GSE65904 was employed to corroborate the validity of the FRGSig. Both univariate and multivariate Cox proportional hazard regression analyses were utilized to construct a FRGSig composed of five FRGs. mRNA expression and immunohistochemistry (IHC) analysis demonstrated that the expression of FRGSig genes varied between tumor and normal tissues. According to Kaplan-Meier analysis, patients with elevated FRGsig scores faced a worse prognosis. The predictive accuracy of FRGSig was evaluated using the time-dependent receiver operating characteristic curve (ROC), with the area under the curve (AUC) values for 1, 3, and 5 OS at 0.682, 0.711, 0.735 in the TCGA cohort, and 0.662, 0.695, 0.712 in the validation dataset, respectively. Univariate and multivariate Cox regression analyses demonstrated that FRGSig served as an independent prognostic factor. Further analysis revealed a significant relationship between FRGSig and Tumor Mutational Burden (TMB) as well as immune infiltration levels. Gene set enrichment analysis (GSEA) disclosed functional disparities between high- and low-risk groups, suggesting that immune checkpoint-related pathways could be instrumental in the improved prognosis of the low-risk group. Taken together, the FRGSig has potential guidance for prognosis prediction and clinical treatment of CM.
Collapse
Affiliation(s)
- Wenna Guo
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xue Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yanting Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Hongtao Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Shanshan Ma
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Corresponding author.
| | - Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Corresponding author.
| |
Collapse
|
8
|
Gu X, Cai L, Luo Z, Shi L, Peng Z, Sun Y, Chen J. Identification and validation of a muscle failure index to predict prognosis and immunotherapy in lung adenocarcinoma through integrated analysis of bulk and single-cell RNA sequencing data. Front Immunol 2023; 13:1057088. [PMID: 36733390 PMCID: PMC9888242 DOI: 10.3389/fimmu.2022.1057088] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/28/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND It was previously reported that the production of exerkines is positively associated with the beneficial effects of exercise in lung adenocarcinoma (LUAD) patients. This study proposes a novel scoring system based on muscle failure-related genes, to assist in clinical decision making. METHODS A comprehensive analysis of bulk and single cell RNA sequencing (scRNA-seq) of early, advanced and brain metastatic LUAD tissues and normal lung tissues was performed to identify muscle failure-related genes in LUAD and to determine the distribution of muscle failure-related genes in different cell populations. A novel scoring system, named MFI (Muscle failure index), was developed and validated. The differences in biological functions, immune infiltration, genomic alterations, and clinical significance of different subtypes were also investigated. RESULTS First, we conducted single cell analysis on the dataset GSE131907 and identified eight cell subpopulations. We found that four muscle failure-related genes (BDNF, FNDC5, IL15, MSTN) were significantly increased in tumor cells. In addition, IL15 was widely distributed in the immune cell population. And we have validated it in our own clinical cohort. Then we created the MFI model based on 10 muscle failure-related genes using the LASSO algorithm, and MFI remained an independent prognostic factor of OS in both the training and validation cohorts. Moreover, we generated MFI in the single-cell dataset, in which cells with high MFI received and sent more signals compared to those with low MFI. Biological function analysis of both subtypes revealed stronger anti-tumor immune activity in the low MFI group, while tumor cells with high MFI had stronger metabolic and proliferative activity. Finally, we systematically assessed the immune cell activity and immunotherapy responses in LUAD patients, finding that the low MFI group was more sensitive to immunotherapy. CONCLUSION Overall, our study can improve the understanding of the role of muscle failure-related genes in tumorigenesis and we constructed a reliable MFI model for predicting prognosis and guiding future clinical decision making.
Collapse
Affiliation(s)
- Xuyu Gu
- School of Medicine, Southeast University, Nanjing, China
| | - Lubing Cai
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Luze Shi
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhen Peng
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yaying Sun
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiwu Chen
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
9
|
Gómez YM, Gallardo DI, Bourguignon M, Bertolli E, Calsavara VF. A general class of promotion time cure rate models with a new biological interpretation. LIFETIME DATA ANALYSIS 2023; 29:66-86. [PMID: 36114312 DOI: 10.1007/s10985-022-09575-3] [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: 10/28/2021] [Accepted: 08/30/2022] [Indexed: 06/15/2023]
Abstract
Over the last decades, the challenges in survival models have been changing considerably and full probabilistic modeling is crucial in many medical applications. Motivated from a new biological interpretation of cancer metastasis, we introduce a general method for obtaining more flexible cure rate models. The proposal model extended the promotion time cure rate model. Furthermore, it includes several well-known models as special cases and defines many new special models. We derive several properties of the hazard function for the proposed model and establish mathematical relationships with the promotion time cure rate model. We consider a frequentist approach to perform inferences, and the maximum likelihood method is employed to estimate the model parameters. Simulation studies are conducted to evaluate its performance with a discussion of the obtained results. A real dataset from population-based study of incident cases of melanoma diagnosed in the state of São Paulo, Brazil, is discussed in detail.
Collapse
Affiliation(s)
- Yolanda M Gómez
- Departamento de Medicina, Facultad de Medicina, Universidad de Atacama, Copiapó, Chile
| | - Diego I Gallardo
- Departamento de Medicina, Facultad de Medicina, Universidad de Atacama, Copiapó, Chile
- Departamento de Matemática, Facultad de Ingeniería, Universidad de Atacama, Copiapó, Chile
| | - Marcelo Bourguignon
- Departamento de Estatística, Universidade Federal do Rio Grande do Norte, Natal, RN, 59078-970, Brazil.
| | - Eduardo Bertolli
- Skin Cancer Department, A.C.Camargo Cancer Center, São Paulo, SP, Brazil
- Oncology Center, Beneficência Portuguesa, São Paulo, SP, Brazil
| | - Vinicius F Calsavara
- Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
10
|
Identification and Validation of Ferroptosis-Related DNA Methylation Signature for Predicting the Prognosis and Guiding the Treatment in Cutaneous Melanoma. Int J Mol Sci 2022; 23:ijms232415677. [PMID: 36555319 PMCID: PMC9778758 DOI: 10.3390/ijms232415677] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Cutaneous melanoma (CM) is one of the most aggressive skin tumors with a poor prognosis. Ferroptosis is a newly discovered form of regulated cell death that is closely associated with cancer development and immunotherapy. The aim of this study was to establish and validate a ferroptosis-related gene (FRG) DNA methylation signature to predict the prognosis of CM patients using data from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) database. A reliable four-FRG DNA methylation prognostic signature was constructed via Cox regression analysis based on TCGA database. Kaplan-Meier analysis showed that patients in the high-risk group tended to have a shorter overall survival (OS) than the low-risk group in both training TCGA and validation GEO cohorts. Time-dependent receiver operating characteristic (ROC) analysis showed the areas under the curve (AUC) at 1, 3, and 5 years were 0.738, 0.730, and 0.770 in TCGA cohort and 0.773, 0.775, and 0.905 in the validation cohort, respectively. Univariate and multivariate Cox regression analyses indicated that the signature was an independent prognostic indicator of OS in patients with CM. Immunogenomic profiling showed the low-risk group of patients had a higher immunophenoscore, and most immune checkpoints were negatively associated with the risk signature. Functional enrichment analysis revealed that immune response and immune-related pathways were enriched in the low-risk group. In conclusion, we established and validated a four-FRG DNA methylation signature that independently predicts prognosis in CM patients. This signature was strongly correlated with the immune landscape, and may serve as a biomarker to guide clinicians in making more precise and personalized treatment decisions for CM patients.
Collapse
|
11
|
Ladányi A, Rásó E, Barbai T, Vízkeleti L, Puskás LG, Kovács SA, Győrffy B, Tímár J. Identification of a Tumor Cell Associated Type I IFN Resistance Gene Expression Signature of Human Melanoma, the Components of Which Have a Predictive Potential for Immunotherapy. Int J Mol Sci 2022; 23:2704. [PMID: 35269844 PMCID: PMC8911010 DOI: 10.3390/ijms23052704] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/24/2022] [Accepted: 02/26/2022] [Indexed: 02/04/2023] Open
Abstract
We developed a human melanoma model using the HT168-M1 cell line to induce IFN-α2 resistance in vitro (HT168-M1res), which was proven to be maintained in vivo in SCID mice. Comparing the mRNA profile of in vitro cultured HT168-M1res cells to its sensitive counterpart, we found 79 differentially expressed genes (DEGs). We found that only a 13-gene core of the DEGs was stable in vitro and only a 4-gene core was stable in vivo. Using an in silico cohort of IFN-treated melanoma tissues, we validated a differentially expressed 9-gene core of the DEGs. Furthermore, using an in silico cohort of immune checkpoint inhibitor (ICI)-treated melanoma tissues, we tested the predictive power of the DEGs for the response rate. Analysis of the top four upregulated and top four downregulated genes of the DEGs identified WFDC1, EFNA3, DDX10, and PTBP1 as predictive genes, and analysis of the "stable" genes of DEGs for predictive potential of ICI response revealed another 13 genes, out of which CDCA4, SOX4, DEK, and HSPA1B were identified as IFN-regulated genes. Interestingly, the IFN treatment associated genes and the ICI-therapy predictive genes overlapped by three genes: WFDC1, BCAN, and MT2A, suggesting a connection between the two biological processes.
Collapse
Affiliation(s)
- Andrea Ladányi
- Department of Surgical and Molecular Pathology, National Institute of Oncology, 1122 Budapest, Hungary;
| | - Erzsébet Rásó
- 2nd Department of Pathology, Semmelweis University, 1091 Budapest, Hungary; (E.R.); (T.B.); (L.V.)
| | - Tamás Barbai
- 2nd Department of Pathology, Semmelweis University, 1091 Budapest, Hungary; (E.R.); (T.B.); (L.V.)
| | - Laura Vízkeleti
- 2nd Department of Pathology, Semmelweis University, 1091 Budapest, Hungary; (E.R.); (T.B.); (L.V.)
| | | | - Szonja A. Kovács
- Department of Bioinformatics, Semmelweis University, 1094 Budapest, Hungary; (S.A.K.); (B.G.)
| | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, 1094 Budapest, Hungary; (S.A.K.); (B.G.)
- Research Centre for Natural Sciences, Oncology Biomarkers Research Group, Institute of Enzymology, Eötvös Loránd Research Network, 1117 Budapest, Hungary
| | - József Tímár
- 2nd Department of Pathology, Semmelweis University, 1091 Budapest, Hungary; (E.R.); (T.B.); (L.V.)
| |
Collapse
|
12
|
Rafei-Shamsabadi D, Lehr S, Behrens M, Meiss F. Additive Intralesional Interleukin-2 Improves Progression-Free Survival in a Distinct Subgroup of Melanoma Patients with Prior Progression under Immunotherapy. Cancers (Basel) 2022; 14:cancers14030540. [PMID: 35158808 PMCID: PMC8833633 DOI: 10.3390/cancers14030540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/05/2022] [Accepted: 01/19/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Despite the immense progress in systemic treatment of advanced unresectable melanoma with immunotherapies, there is still an unmet need for patients showing primary resistance. We show that addition of intratumoral injections of the immune modulator Interleukin-2 can overcome primary resistance in a distinct subset of patients and this effect is associated with an intratumoral increase of certain inflammatory cells supporting the paradigm of turning a “cold” into a “hot” tumor. Abstract A considerable amount of melanoma patients show primary resistance to PD-1 and CTLA-4 inhibitors. We have previously reported a beneficial role of intralesional Interleukin-2 (IL-2) in 9 melanoma patients developing new locoregional metastases under immunotherapy. We have now expanded this retrospective cohort to 27 patients. Patients were evaluated for their tumor characteristics, treatment response and progression-free and overall survival (PFS/OS). In 16 patients, tumor biopsies before and under IL-2 treatment were evaluated for immune markers. The median follow-up time was 16 (1–59) months from start of IL-2 treatment. Treatment response of locoregional metastases was seen in 74% of all patients and response of distant organ metastases in 37% of stage IV patients, respectively. A prolonged PFS and OS was significantly associated with absence of active distant metastases (p = 0.008), response of locoregional metastases (p = 0.002), increase of absolute eosinophil count (AEC) (p < 0.001) and an influx of CD8+ tumor infiltrating lymphocytes (TILs) (p = 0.003). Additional intralesional treatment with IL-2 in patients with locoregional progression under immunotherapy is a well-tolerated, easily feasible therapeutic option especially in patients lacking active distant metastases. A careful patient selection can lead to an improved PFS and OS.
Collapse
Affiliation(s)
- David Rafei-Shamsabadi
- Department of Dermatology and Venereology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 7, 79104 Freiburg, Germany; (D.R.-S.); (S.L.)
| | - Saskia Lehr
- Department of Dermatology and Venereology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 7, 79104 Freiburg, Germany; (D.R.-S.); (S.L.)
| | - Max Behrens
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Center—University of Freiburg, Zinkmattenstr. 6a, 79108 Freiburg, Germany;
| | - Frank Meiss
- Department of Dermatology and Venereology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 7, 79104 Freiburg, Germany; (D.R.-S.); (S.L.)
- Correspondence:
| |
Collapse
|
13
|
Quiroz TS, Heras FL. Medicina de precisión: rol de la anatomía patológica en diagnóstico y tratamiento personalizado. REVISTA MÉDICA CLÍNICA LAS CONDES 2022. [DOI: 10.1016/j.rmclc.2022.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
14
|
Becker AL, Carpenter EL, Slominski AT, Indra AK. The Role of the Vitamin D Receptor in the Pathogenesis, Prognosis, and Treatment of Cutaneous Melanoma. Front Oncol 2021; 11:743667. [PMID: 34692525 PMCID: PMC8526885 DOI: 10.3389/fonc.2021.743667] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/15/2021] [Indexed: 01/08/2023] Open
Abstract
Melanoma is the malignant transformation of melanocytes and represents the most lethal form of skin cancer. While early-stage melanoma localized to the skin can be cured with surgical excision, metastatic melanoma often requires a multi-pronged approach and even then can exhibit treatment resistance. Understanding the molecular mechanisms involved in the pathogenesis of melanoma could lead to novel diagnostic, prognostic, and therapeutic strategies to ultimately decrease morbidity and mortality. One emerging candidate that may have value as both a prognostic marker and in a therapeutic context is the vitamin D receptor (VDR). VDR is a nuclear steroid hormone receptor activated by 1,25 dihydroxy-vitamin D3 [calcitriol, 1,25(OH)2D3]. While 1,25 dihydroxy-vitamin D3 is typically thought of in relation to calcium metabolism, it also plays an important role in cell proliferation, differentiation, programmed-cell death as well as photoprotection. This review discusses the role of VDR in the crosstalk between keratinocytes and melanocytes during melanomagenesis and summarizes the clinical data regarding VDR polymorphisms, VDR as a prognostic marker, and potential uses of vitamin D and its analogs as an adjuvant treatment for melanoma.
Collapse
Affiliation(s)
- Alyssa L. Becker
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU, Corvallis, OR, United States
- John A. Burns School of Medicine at the University of Hawai’i at Mānoa, Honolulu, HI, United States
| | - Evan L. Carpenter
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU, Corvallis, OR, United States
| | - Andrzej T. Slominski
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, United States
- Cancer Chemoprevention Program, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Arup K. Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU, Corvallis, OR, United States
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR, United States
- Department of Biochemistry and Biophysics, Oregon State University (OSU), Corvallis, OR, United States
- Linus Pauling Science Center, Oregon State University (OSU), Corvallis, OR, United States
- Department of Dermatology, Oregon Health & Science University (OHSU), Portland, OR, United States
| |
Collapse
|
15
|
Lee MH. Harness the functions of gut microbiome in tumorigenesis for cancer treatment. Cancer Commun (Lond) 2021; 41:937-967. [PMID: 34355542 PMCID: PMC8504147 DOI: 10.1002/cac2.12200] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/16/2021] [Indexed: 11/08/2022] Open
Abstract
It has been shown that gut microbiota dysbiosis leads to physiological changes and links to a number of diseases, including cancers. Thus, many cancer categories and treatment regimens should be investigated in the context of the microbiome. Owing to the availability of metagenome sequencing and multiomics studies, analyses of species characterization, host genetic changes, and metabolic profile of gut microbiota have become feasible, which has facilitated an exponential knowledge gain about microbiota composition, taxonomic alterations, and host interactions during tumorigenesis. However, the complexity of the gut microbiota, with a plethora of uncharacterized host‐microbe, microbe‐microbe, and environmental interactions, still contributes to the challenge of advancing our knowledge of the microbiota‐cancer interactions. These interactions manifest in signaling relay, metabolism, immunity, tumor development, genetic instability, sensitivity to cancer chemotherapy and immunotherapy. This review summarizes current studies/molecular mechanisms regarding the association between the gut microbiota and the development of cancers, which provides insights into the therapeutic strategies that could be harnessed for cancer diagnosis, treatment, or prevention.
Collapse
Affiliation(s)
- Mong-Hong Lee
- Research Institute of Gastroenterology, Sun Yat-sen University, Guangzhou, Guangdong, 510020, P. R. China.,Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510020, P. R. China
| |
Collapse
|