1
|
Palkovsky M, Modrackova N, Neuzil-Bunesova V, Liberko M, Soumarova R. The Bidirectional Impact of Cancer Radiotherapy and Human Microbiome: Microbiome as Potential Anti-tumor Treatment Efficacy and Toxicity Modulator. In Vivo 2025; 39:37-54. [PMID: 39740900 PMCID: PMC11705129 DOI: 10.21873/invivo.13803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/02/2024] [Accepted: 10/14/2024] [Indexed: 01/02/2025]
Abstract
Microbiome and radiotherapy represent bidirectionally interacting entities. The human microbiome has emerged as a pivotal modulator of the efficacy and toxicity of radiotherapy; however, a reciprocal effect of radiotherapy on microbiome composition alterations has also been observed. This review explores the relationship between the microbiome and extracranial solid tumors, particularly focusing on the bidirectional impact of radiotherapy on organ-specific microbiome. This article aims to provide a systematic review on the radiotherapy-induced microbial alteration in-field as well as in distant microbiomes. In this review, particular focus is directed to the oral and gut microbiome, its role in the development and progression of cancer, and how it is altered throughout radiotherapy. This review concludes with recommendations for future research, such as exploring microbiome modification to optimize radiotherapy-induced toxicities or enhance its anti-cancer effects.
Collapse
Affiliation(s)
- Martin Palkovsky
- Department of Oncology, University Hospital Kralovske Vinohrady, Prague, Czech Republic;
- Charles University, Third Faculty of Medicine, Department of Oncology, Prague, Czech Republic
| | - Nikol Modrackova
- Czech University of Life Sciences Prague, Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Prague, Czech Republic
| | - Vera Neuzil-Bunesova
- Czech University of Life Sciences Prague, Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Prague, Czech Republic
| | - Marian Liberko
- Department of Oncology, University Hospital Kralovske Vinohrady, Prague, Czech Republic
- Charles University, Third Faculty of Medicine, Department of Oncology, Prague, Czech Republic
| | - Renata Soumarova
- Department of Oncology, University Hospital Kralovske Vinohrady, Prague, Czech Republic
- Charles University, Third Faculty of Medicine, Department of Oncology, Prague, Czech Republic
| |
Collapse
|
2
|
Ohsawa M, Nishi H, Emi M, Yoshikawa T, Hamai Y, Ibuki Y, Kurokawa T, Hirohata R, Kitasaki N, Kawada-Matsuo M, Komatsuzawa H, Kawaguchi H, Okada M. Impact of Fusobacterium nucleatum in the treatment of cancer, including radiotherapy and its future potential in esophageal cancer. JOURNAL OF RADIATION RESEARCH 2024; 65:i126-i134. [PMID: 39679879 DOI: 10.1093/jrr/rrae061] [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: 12/02/2023] [Revised: 05/09/2024] [Indexed: 12/17/2024]
Abstract
Despite advances in multimodality therapy, including surgery, chemotherapy, radiation therapy and chemoradiation, the fatality rate for esophageal cancer remains high. Specifically, Fusobacterium nucleatum, due to its aggregation capacity, has shown a tendency to form biofilms. The biofilm-forming capabilities of microbial communities are of utmost importance in the context of cancer treatment, as they have been shown to drive significant losses in the efficaciousness of various cancer treatments. Therefore, elucidating the dynamics of F. nucleatum will be important for the development of effective treatments for esophageal cancer. Therefore, this review summarizes the current knowledge of F. nucleatum, its involvement in cancer and its impact on chemotherapy and radiation therapy. In conclusion, further research on the role of F. nucleatum is essential for the continued advancement of the treatment of esophageal cancer and patient care.
Collapse
Affiliation(s)
- Manato Ohsawa
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Hiromi Nishi
- Department of General Dentistry, Hiroshima University Hospital, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Manabu Emi
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Toru Yoshikawa
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Yoichi Hamai
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Yuta Ibuki
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Tomoaki Kurokawa
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Ryosuke Hirohata
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Nao Kitasaki
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Miki Kawada-Matsuo
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Hitoshi Komatsuzawa
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Hiroyuki Kawaguchi
- Department of General Dentistry, Hiroshima University Hospital, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Morihito Okada
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| |
Collapse
|
3
|
Shaikh FY, Lee S, White JR, Zhao Y, Ferri JT, Pereira G, Landon BV, Ke S, Hu C, Feliciano JL, Hales RK, Voong KR, Battafarano RJ, Yang SC, Broderick S, Ha J, Thompson E, Shin EJ, Bartlett DL, Weksler B, Pardoll DM, Anagnostou V, Lam VK, Zaidi AH, Kelly RJ, Sears CL. Fecal Microbiome Composition Correlates with Pathologic Complete Response in Patients with Operable Esophageal Cancer Treated with Combined Chemoradiotherapy and Immunotherapy. Cancers (Basel) 2024; 16:3644. [PMID: 39518082 PMCID: PMC11545537 DOI: 10.3390/cancers16213644] [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: 09/17/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Preclinical and clinical data indicate that chemoradiotherapy (CRT) in combination with checkpoint inhibitors may prime an anti-tumor immunological response in esophageal cancer. However, responses to neoadjuvant therapy can vary widely and the key biomarkers to determine response remain poorly understood. The fecal microbiome is a novel and potentially modifiable biomarker of immunotherapy response, and both fecal and tumor microbes have been found to associate with outcomes in esophageal cancer. Methods: Fecal and tumor samples were collected from patients with stage II-III resectable esophageal or gastroesophageal junction carcinoma treated with neoadjuvant immune checkpoint inhibitors (ICIs) plus CRT prior to surgical resection. Microbiome profiles were analyzed by 16S rRNA amplicon sequencing and taxonomic data were integrated with fecal metabolite analysis to assess microbial function. Results: The fecal microbiome of patients with pathological complete response (PCR) grouped in distinct clusters compared to patients with residual viable tumor (RVT) by Bray-Curtis diversity metric. Integrated taxonomic and metabolomic analysis of fecal samples identified a sphingolipid and primary bile acid as enriched in the PCR, the levels of which correlated with several bacterial species: Roseburis inulinivorans, Ruminococcus callidus, and Fusicantenibacter saccharivorans. Analysis of the tumor microbiome profiles identified several bacterial genera previously associated with esophageal tumors, including Streptococcus and Veillonella. Conclusions: These results further characterize the fecal and tumor microbiome of patients with operable esophageal cancer and identify specific microbes and metabolites that may help elucidate how microbes contribute to tumor response with neoadjuvant CRT combined with ICI.
Collapse
Affiliation(s)
- Fyza Y. Shaikh
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Bloomberg-Kimmel Institute of Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Seoho Lee
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | - Yujie Zhao
- Department of Biomedical Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA
| | - Jacqueline T. Ferri
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Gavin Pereira
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Blair V. Landon
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Suqi Ke
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Chen Hu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Josephine L. Feliciano
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Russell K. Hales
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Radiation Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - K. Ranh Voong
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Radiation Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard J. Battafarano
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Stephen C. Yang
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Stephen Broderick
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jinny Ha
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elizabeth Thompson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Eun J. Shin
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - David L. Bartlett
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, PA 15212, USA
| | - Benny Weksler
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, PA 15212, USA
| | - Drew M. Pardoll
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Bloomberg-Kimmel Institute of Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Valsamo Anagnostou
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Bloomberg-Kimmel Institute of Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Vincent K. Lam
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ali H. Zaidi
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, PA 15212, USA
| | - Ronan J. Kelly
- The Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX 75246, USA
| | - Cynthia L. Sears
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Bloomberg-Kimmel Institute of Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
4
|
Dou Z, Ai C, Zhang J, Li K, Jiang M, Wu X, Zhao C, Li Z, Zhang L. The intra-tumoral microbiome as a potential biomarker of response to external beam radiation therapy in cervical cancer. J Transl Med 2024; 22:972. [PMID: 39468630 PMCID: PMC11514760 DOI: 10.1186/s12967-024-05774-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND We aimed to determine the potential predictive value of the intra-tumoral microbiome as a marker of the response to external beam radiation therapy (EBRT) in cervical cancer (CC). METHODS A prospective longitudinal trial of 36 CC patients receiving pelvic radiotherapy was designed to investigate microbial characteristic signatures and diversity (alpha and beta) of multiple sites (tumor, vaginal, gut, urethral, and oral) in the superior response (SR) and inferior response (IR) groups of CC patients by 16S rRNA sequencing. Utilized the least absolute shrinkage and selection operator (LASSO) logistic regression method to analyze clinicopathological factors that potentially influenced the efficacy of EBRT. LEfSe analysis highlighted the microbiome features that best distinguished the categorized patient samples. Selected parameters were validated with Spearman correlation analysis, receiver operating characteristic (ROC) area under the curve (AUC) analysis and Kaplan-Meier survival analysis. RESULTS Firstly, in our cohort, LASSO logistic regression analysis revealed no association between clinicopathological factors and EBRT efficacy. Subsequently, we employed 16S rRNA sequencing to compare microbiome differences across multiple sites and their correlations with major clinicopathological factors. We discovered that the intra-tumoral microbiome was independent of clinicopathologic features and represented the most direct and reliable reflection of the microbial differences between the SR and IR groups. We found lower alpha diversity in the tumor microbiome of SR group and identified the most relevant microbiome taxa (Bifidobacteriaceae, Beijerinckiaceae, and Orbaceae) associated with the efficacy of the response to EBRT in CC patients. We then conducted ROC analysis, finding that specific microbial taxa had an AUC of 0.831 (95% CI, 0.667-0.995), indicating the potential of these taxa as biomarkers for predicting EBRT efficacy. Kaplan-Meier survival analysis showed a better prognosis for patients with lower alpha diversity and higher relative abundance of Bifidobacteriaceae. CONCLUSIONS Our data suggested that intra-tumoral specific microbiome taxa and lower alpha diversity may play an important role in the CC patient sensitivity to EBRT and offer novel potential biomarkers for predicting the response to EBRT efficacy.
Collapse
Affiliation(s)
- Zhongyan Dou
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), 519 Kunzhou Road, Kunming, 650118, China
| | - Conghui Ai
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), 519 Kunzhou Road, Kunming, 650118, China
| | - Jinping Zhang
- Department of Medical Administration, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), 519 Kunzhou Road, Kunming, 650118, China
| | - Kangming Li
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), 519 Kunzhou Road, Kunming, 650118, China
| | - Meiping Jiang
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), 519 Kunzhou Road, Kunming, 650118, China
| | - Xingrao Wu
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), 519 Kunzhou Road, Kunming, 650118, China
| | - Chunfang Zhao
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), 519 Kunzhou Road, Kunming, 650118, China
| | - Zheng Li
- Department of Gynecologic Oncology, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), 519 Kunzhou Road, Kunming, 650118, China.
| | - Lan Zhang
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), 519 Kunzhou Road, Kunming, 650118, China.
| |
Collapse
|
5
|
He Y, Li XY, Hu AQ, Qian D. Salivary microbiome is associated with the response to chemoradiotherapy in initially inoperable patients with esophageal squamous cell carcinoma. J Oral Microbiol 2024; 16:2359887. [PMID: 38813524 PMCID: PMC11134033 DOI: 10.1080/20002297.2024.2359887] [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: 02/02/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024] Open
Abstract
Background The salivary microbiome may interact with chemoradiotherapy through dynamic changes in microbial composition and systemic immunity. We aimed to explore the association between the salivary microbiome and response to chemoradiotherapy in initially inoperable patients with local advanced esophageal squamous cell carcinoma (LAESCC). Methods Salivary and peripheral blood samples were collected before and after chemoradiotherapy. The microbiome and metabolic pathways were analyzed by 16S ribosomal RNA sequencing and liquid chromatography tandem mass spectrometry/Mass spectrometry analyses. Results The salivary microbiome exhibited characteristic variations between patients and healthy controls. A significant correlation was found between Prevotella_salivae, Saccharibacteria_TM7_G3_bacterium_HMT_351, and Veillonellaceae_G1_bacterium_HMT_129 and pathological complete response (pCR) in initially inoperable patients who underwent surgery. The PICRUSt suggested that immune diseases and cell motility were different in tumor compared to normal groups. KEGG enrichment analysis showed enriched lipid metabolism, signal transduction, and membrane transport in the tumor group. CD3+CD8 T cells, IL6, IL10, and IFNγ exhibited an increasing trend during the treatment process of chemoradiotherapy. Conclusions Our study demonstrated that variations in specific saliva taxa associated with host immunomodulatory cells and cytokines could be promising for early efficacy prediction of chemoradiotherapy in initially inoperable patients with LAESCC.
Collapse
Affiliation(s)
- Yuan He
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiao-Yang Li
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - An-Qi Hu
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Dong Qian
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
6
|
Wang K, Wang J, Chen Y, Long H, Pan W, Liu Y, Xu MY, Guo Q. Causal relationship between gut microbiota and risk of esophageal cancer: evidence from Mendelian randomization study. Aging (Albany NY) 2024; 16:3596-3611. [PMID: 38364235 PMCID: PMC10929825 DOI: 10.18632/aging.205547] [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: 10/19/2023] [Accepted: 01/11/2024] [Indexed: 02/18/2024]
Abstract
BACKGROUND The causative implications remain ambiguous. Consequently, this study aims to evaluate the putative causal relationship between gut microbiota and Esophageal cancer (EC). METHODS The genome-wide association study (GWAS) pertaining to the microbiome, derived from the MiBioGen consortium-which consolidates 18,340 samples across 24 population-based cohorts-was utilized as the exposure dataset. Employing the GWAS summary statistics specific to EC patients sourced from the GWAS Catalog and leveraging the two-sample Mendelian randomization (MR) methodology, the principal analytical method applied was the inverse variance weighted (IVW) technique. Cochran's Q statistic was utilized to discern heterogeneity inherent in the data set. Subsequently, a reverse MR analysis was executed. RESULTS Findings derived from the IVW technique elucidated that the Family Porphyromonadaceae (P = 0.048) and Genus Candidatus Soleaferrea (P = 0.048) function as deterrents against EC development. In contrast, the Genus Catenibacterium (P = 0.044), Genus Eubacterium coprostanoligenes group (P = 0.038), Genus Marvinbryantia (P = 0.049), Genus Ruminococcaceae UCG010 (P = 0.034), Genus Ruminococcus1 (P = 0.047), and Genus Sutterella (P = 0.012) emerged as prospective risk contributors for EC. To assess reverse causal effect, we used EC as the exposure and the gut microbiota as the outcome, and this analysis revealed associations between EC and seven different types of gut microbiota. The robustness of the MR findings was substantiated through comprehensive heterogeneity and pleiotropy evaluations. CONCLUSIONS This research identified certain microbial taxa as either protective or detrimental elements for EC, potentially offering valuable biomarkers for asymptomatic diagnosis and prospective therapeutic interventions for EC.
Collapse
Affiliation(s)
- Kui Wang
- Department of Gastroenterology, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
- Medical School, Kunming University of Science and Technology, Kunming 650500, Yunnan Province, China
| | - Jiawei Wang
- Department of Critical Care Medicine, Jieyang Third People’s Hospital, Jieyang 515500, Guangdong Province, China
| | - Yuhua Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Huan Long
- Department of Gastroenterology, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
- Medical School, Kunming University of Science and Technology, Kunming 650500, Yunnan Province, China
| | - Wei Pan
- Cardiology Department, Geriatrics Department, Foshan Women and Children Hospital, Foshan 528000, Guangdong, China
| | - Yunfei Liu
- University Munich, Munich D-81377, Germany
| | - Ming-Yi Xu
- Department of Gastroenterology, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai 310115, China
| | - Qiang Guo
- Department of Gastroenterology, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| |
Collapse
|