1
|
Ataollahi H, Hedayati M, Zia-Jahromi N, Daneshpour M, Siadat SD. Investigating the role of the intratumoral microbiome in thyroid cancer development and progression. Crit Rev Oncol Hematol 2024; 204:104545. [PMID: 39476992 DOI: 10.1016/j.critrevonc.2024.104545] [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: 07/11/2024] [Revised: 10/14/2024] [Accepted: 10/24/2024] [Indexed: 11/04/2024] Open
Abstract
The intratumoral microbiome (ITM) is in the spotlight due to its possible contribution to the initiation, progression, and invasion of a wide range of cancers. Its precise contribution to cancer tumorigenesis is still elusive, though. Thyroid cancer(TC), the ninth leading cause of cancer globally and the most prevalent endocrine malignancy with a rapidly rising incidence among all cancers, has attracted much attention nowadays. Still, the association between the tumor's microbiome and TC progression and development is an evolving area of investigation with significant consequences for disease understanding and intervention. Therefore, this review offers an appropriate perspective on this emerging concept in TC based on prior studies on the ITM among the most common tumors worldwide, concentrating on TC. Moreover, information on the origin of the ITM and practical methods can pave the way for researchers to opt for the most appropriate method for further investigations on the ITM more accurately.
Collapse
Affiliation(s)
- Hanieh Ataollahi
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No 23, Shahid Arabi St.Yemen St, Velenjak, PO Box:19395-4763, Tehran, Iran.
| | - Noosha Zia-Jahromi
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Maryam Daneshpour
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No 23, Shahid Arabi St.Yemen St, Velenjak, PO Box:19395-4763, Tehran, Iran
| | - Seyed Davar Siadat
- Microbiology Research Center(MRC), Pasteur Institute of Iran, Tehran, Iran; Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
2
|
Zeng X, Ma Q, Huang CX, Xiao JJ, Fu X, Ren YF, Qu YL, Xiang HX, Lei M, Zheng RY, Zhong Y, Xiao P, Zhuang X, You FM, He JW. Diagnostic potential of salivary microbiota in persistent pulmonary nodules: identifying biomarkers and functional pathways using 16S rRNA sequencing and machine learning. J Transl Med 2024; 22:1079. [PMID: 39609902 PMCID: PMC11603953 DOI: 10.1186/s12967-024-05802-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/23/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND The aim of this study was to explore the microbial variations and biomarkers in the oral environment of patients with persistent pulmonary nodules (pPNs) and to reveal the potential biological functions of the salivary microbiota in pPNs. MATERIALS AND METHODS This study included a total of 483 participants (141 healthy controls and 342 patients with pPNs) from June 2022 and January 2024. Saliva samples were subjected to sequencing of the V3-V4 region of the 16S rRNA gene to assess microbial diversity and differential abundance. Seven advanced machine learning algorithms (logistic regression, support vector machine, multi-layer perceptron, naïve Bayes, random forest, gradient boosting decision tree, and LightGBM) were utilized to evaluate performance and identify key microorganisms, with fivefold cross-validation employed to ensure robustness. The Shapley Additive exPlanations (SHAP) algorithm was employed to explain the contribution of these core microbiotas to the predictive model. Additionally, the PICRUSt2 algorithm was used to predict the microbial functions. RESULTS The salivary microbial composition in pPNs group showed significantly lower α- and β-diversity compared to healthy controls. A high-accuracy LightGBM model was developed, identifying six core genera-Fusobacterium, Solobacterium, Actinomyces, Porphyromonas, Atopobium, and Peptostreptococcus-as pPNs biomarkers. Additionally, a visualization pPNs risk prediction system was developed. The immune responses and metabolic activities differences in salivary microbiota between the patients with pPNs and healthy controls were revealed. CONCLUSIONS This study highlights the potential clinical applications of the salivary microbiota for enable earlier detection and targeted interventions, offering significant promise for advancing clinical management and improving patient outcomes in pPNs.
Collapse
Affiliation(s)
- Xiao Zeng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Qiong Ma
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Chun-Xia Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Jun-Jie Xiao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Xi Fu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Yi-Feng Ren
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Yu-Li Qu
- College of Artificial Intelligence, Xi'an Jiaotong University, Xian, 710061, Shanxi Province, China
| | - Hong-Xia Xiang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Mao Lei
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Ru-Yi Zheng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Yang Zhong
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Ping Xiao
- Department of Thoracic Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610042, Sichuan Province, China
| | - Xiang Zhuang
- Department of Thoracic Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610042, Sichuan Province, China
| | - Feng-Ming You
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China.
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China.
| | - Jia-Wei He
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China.
| |
Collapse
|
3
|
Lee YH, Jung J, Hong JY. Oral Microbial Changes in Oral Squamous Cell Carcinoma: Focus on Treponema denticola, Lactobacillus casei, and Candida albicans. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1753. [PMID: 39596938 PMCID: PMC11596053 DOI: 10.3390/medicina60111753] [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: 09/04/2024] [Revised: 09/24/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024]
Abstract
Background and Objectives: In this study, we aimed to explore the oral bacteria and fungi that can help discern oral squamous cell carcinoma (OSCC) and investigate the correlations between multiple key pathogens. Materials and Methods: Twelve participants (8 females and 4 males; mean age, 54.33 ± 20.65 years) were prospectively recruited into three groups: Group 1: healthy control, Group 2: patients with stomatitis, and Group 3: patients with OSCC, with 4 individuals in each group. Unstimulated whole saliva samples from these participants were analyzed using real-time PCR to assess the presence and abundance of 14 major oral bacterial species and Candida albicans. Results: The analysis revealed significant differences for certain microorganisms, namely, Treponema denticola (T. denticola), Lactobacillus casei (L. casei), and Candida albicans. T. denticola was most abundant in the OSCC group (5,358,692.95 ± 3,540,767.33), compared to the stomatitis (123,355.54 ± 197,490.86) and healthy control (9999.21 ± 11,998.40) groups. L. casei was undetectable in the healthy control group but was significantly more abundant in the stomatitis group (1653.94 ± 2981.98) and even higher in the OSCC group (21,336.95 ± 9258.79) (p = 0.001). A similar trend was observed for C. albicans, with DNA copy numbers rising from the healthy control (464.29 ± 716.76) to the stomatitis (1861.30 ± 1206.15) to the OSCC group (9347.98 ± 5128.54) (p = 0.006). The amount of T. denticola was positively correlated with L. casei (r = 0.890, p < 0.001) and C. albicans (r = 0.724, p = 0.008). L. casei's DNA copy number was strongly correlated with C. albicans (r = 0.931, p < 0.001). These three oral microbes exhibited strong positive correlations with each other and had various direct or indirect relationships with other species. Conclusions: In the OSCC group, T. denticola, L. casei, and C. albicans exhibited strong positive correlations with one another, further emphasizing the need for a deeper understanding of the complex microbial interactions in the OSCC environment.
Collapse
Affiliation(s)
- Yeon-Hee Lee
- Department of Orofacial Pain and Oral Medicine, Kyung Hee University Dental Hospital, #613 Hoegi-dong, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Junho Jung
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyung Hee University, Dongdaemun-gu, Seoul 02447, Republic of Korea;
| | - Ji-Youn Hong
- Department of Periodontology, Periodontal-Implant Clinical Research Institute, School of Dentistry, Kyung Hee University, Dongdaemun-gu, Seoul 02447, Republic of Korea;
| |
Collapse
|
4
|
Hossain SM, Ly K, Sung YJ, Braithwaite A, Li K. Immune Checkpoint Inhibitor Therapy for Metastatic Melanoma: What Should We Focus on to Improve the Clinical Outcomes? Int J Mol Sci 2024; 25:10120. [PMID: 39337605 PMCID: PMC11432671 DOI: 10.3390/ijms251810120] [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: 06/05/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have transformed cancer treatment by enhancing anti-tumour immune responses, demonstrating significant efficacy in various malignancies, including melanoma. However, over 50% of patients experience limited or no response to ICI therapy. Resistance to ICIs is influenced by a complex interplay of tumour intrinsic and extrinsic factors. This review summarizes current ICIs for melanoma and the factors involved in resistance to the treatment. We also discuss emerging evidence that the microbiota can impact ICI treatment outcomes by modulating tumour biology and anti-tumour immune function. Furthermore, microbiota profiles may offer a non-invasive method for predicting ICI response. Therefore, future research into microbiota manipulation could provide cost-effective strategies to enhance ICI efficacy and improve outcomes for melanoma patients.
Collapse
Affiliation(s)
- Sultana Mehbuba Hossain
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Kevin Ly
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Yih Jian Sung
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Antony Braithwaite
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Kunyu Li
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| |
Collapse
|
5
|
Peng F, Hu M, Su Z, Hu L, Guo L, Yang K. Intratumoral Microbiota as a Target for Advanced Cancer Therapeutics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405331. [PMID: 39054925 DOI: 10.1002/adma.202405331] [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: 04/15/2024] [Revised: 06/21/2024] [Indexed: 07/27/2024]
Abstract
In recent years, advancements in microbial sequencing technology have sparked an increasing interest in the bacteria residing within solid tumors and its distribution and functions in various tumors. Intratumoral bacteria critically modulate tumor oncogenesis and development through DNA damage induction, chronic inflammation, epigenetic alterations, and metabolic and immune regulation, while also influencing cancer treatment efficacy by affecting drug metabolism. In response to these discoveries, a variety of anti-cancer therapies targeting these microorganisms have emerged. These approaches encompass oncolytic therapy utilizing tumor-associated bacteria, the design of biomaterials based on intratumoral bacteria, the use of intratumoral bacterial components for drug delivery systems, and comprehensive strategies aimed at the eradication of tumor-promoting bacteria. Herein, this review article summarizes the distribution patterns of bacteria in different solid tumors, examines their impact on tumors, and evaluates current therapeutic strategies centered on tumor-associated bacteria. Furthermore, the challenges and prospects for developing drugs that target these bacterial communities are also explored, promising new directions for cancer treatment.
Collapse
Affiliation(s)
- Fei Peng
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Mengyuan Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zhiyue Su
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Lin Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Lingchuan Guo
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Kai Yang
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215123, China
- Key Laboratory of Alkene-carbon Fibres-based Technology & Application for Detection of Major Infectious Diseases, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
6
|
Liu J, Li B, Li L, Ming X, Xu ZP. Advances in Nanomaterials for Immunotherapeutic Improvement of Cancer Chemotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403024. [PMID: 38773882 DOI: 10.1002/smll.202403024] [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: 04/15/2024] [Revised: 05/10/2024] [Indexed: 05/24/2024]
Abstract
Immuno-stimulative effect of chemotherapy (ISECT) is recognized as a potential alternative to conventional immunotherapies, however, the clinical application is constrained by its inefficiency. Metronomic chemotherapy, though designed to overcome these limitations, offers inconsistent results, with effectiveness varying based on cancer types, stages, and patient-specific factors. In parallel, a wealth of preclinical nanomaterials holds considerable promise for ISECT improvement by modulating the cancer-immunity cycle. In the area of biomedical nanomaterials, current literature reviews mainly concentrate on a specific category of nanomaterials and nanotechnological perspectives, while two essential issues are still lacking, i.e., a comprehensive analysis addressing the causes for ISECT inefficiency and a thorough summary elaborating the nanomaterials for ISECT improvement. This review thus aims to fill these gaps and catalyze further development in this field. For the first time, this review comprehensively discusses the causes of ISECT inefficiency. It then meticulously categorizes six types of nanomaterials for improving ISECT. Subsequently, practical strategies are further proposed for addressing inefficient ISECT, along with a detailed discussion on exemplary nanomedicines. Finally, this review provides insights into the challenges and perspectives for improving chemo-immunotherapy by innovations in nanomaterials.
Collapse
Affiliation(s)
- Jie Liu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, St Lucia, QLD, 4072, Australia
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 000000, China
- GoodMedX Tech Limited Company, Hong Kong SAR, 000000, China
| | - Bei Li
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Li Li
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, St Lucia, QLD, 4072, Australia
| | - Xin Ming
- Departments of Cancer Biology and Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, North Carolina, 27157, USA
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, St Lucia, QLD, 4072, Australia
- Institute of Biomedical Health Technology and Engineering, and Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, 518107, China
| |
Collapse
|
7
|
Furuta S. Microbiome-Stealth Regulator of Breast Homeostasis and Cancer Metastasis. Cancers (Basel) 2024; 16:3040. [PMID: 39272898 PMCID: PMC11394247 DOI: 10.3390/cancers16173040] [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: 08/21/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Cumulative evidence attests to the essential roles of commensal microbes in the physiology of hosts. Although the microbiome has been a major research subject since the time of Luis Pasteur and William Russell over 140 years ago, recent findings that certain intracellular bacteria contribute to the pathophysiology of healthy vs. diseased tissues have brought the field of the microbiome to a new era of investigation. Particularly, in the field of breast cancer research, breast-tumor-resident bacteria are now deemed to be essential players in tumor initiation and progression. This is a resurrection of Russel's bacterial cause of cancer theory, which was in fact abandoned over 100 years ago. This review will introduce some of the recent findings that exemplify the roles of breast-tumor-resident microbes in breast carcinogenesis and metastasis and provide mechanistic explanations for these phenomena. Such information would be able to justify the utility of breast-tumor-resident microbes as biomarkers for disease progression and therapeutic targets.
Collapse
Affiliation(s)
- Saori Furuta
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, 2500 MetroHealth Drive, Cleveland, OH 44109, USA;
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
8
|
Bi X, Wang J, Liu C. Intratumoral Microbiota: Metabolic Influences and Biomarker Potential in Gastrointestinal Cancer. Biomolecules 2024; 14:917. [PMID: 39199305 PMCID: PMC11353126 DOI: 10.3390/biom14080917] [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: 07/10/2024] [Revised: 07/21/2024] [Accepted: 07/25/2024] [Indexed: 09/01/2024] Open
Abstract
Gastrointestinal (GI) cancers impose a substantial global health burden, highlighting the necessity for deeper understanding of their intricate pathogenesis and treatment strategies. This review explores the interplay between intratumoral microbiota, tumor metabolism, and major types of GI cancers (including esophageal, gastric, liver, pancreatic, and colorectal cancers), summarizing recent studies and elucidating their clinical implications and future directions. Recent research revealed altered microbial signatures within GI tumors, impacting tumor progression, immune responses, and treatment outcomes. Dysbiosis-induced alterations in tumor metabolism, including glycolysis, fatty acid metabolism, and amino acid metabolism, play critical roles in cancer progression and therapeutic resistance. The integration of molecular mechanisms and potential biomarkers into this understanding further enhances the prognostic significance of intratumoral microbiota composition and therapeutic opportunities targeting microbiota-mediated tumor metabolism. Despite advancements, challenges remain in understanding the dynamic interactions within the tumor microenvironment (TME). Future research directions, including advanced omics technologies and prospective clinical studies, offer promising avenues for precision oncology and personalized treatment interventions in GI cancer. Overall, integrating microbiota-based approaches and molecular biomarkers into GI cancer management holds promise for improving patient outcomes and survival.
Collapse
Affiliation(s)
- Xueyuan Bi
- Department of Pharmacy, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Jihan Wang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China;
| | - Cuicui Liu
- Department of Science and Education, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| |
Collapse
|
9
|
Qin H, Liu J, Qu Y, Li YY, Xu YL, Yan YF. The intratumoral microbiota biomarkers for predicting survival and efficacy of immunotherapy in patients with ovarian serous cystadenocarcinoma. J Ovarian Res 2024; 17:140. [PMID: 38970121 PMCID: PMC11227176 DOI: 10.1186/s13048-024-01464-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/26/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Ovarian serous cystadenocarcinoma, accounting for about 90% of ovarian cancers, is frequently diagnosed at advanced stages, leading to suboptimal treatment outcomes. Given the malignant nature of the disease, effective biomarkers for accurate prediction and personalized treatment remain an urgent clinical need. METHODS In this study, we analyzed the microbial contents of 453 ovarian serous cystadenocarcinoma and 68 adjacent non-cancerous samples. A univariate Cox regression model was used to identify microorganisms significantly associated with survival and a prognostic risk score model constructed using LASSO Cox regression analysis. Patients were subsequently categorized into high-risk and low-risk groups based on their risk scores. RESULTS Survival analysis revealed that patients in the low-risk group had a higher overall survival rate. A nomogram was constructed for easy visualization of the prognostic model. Analysis of immune cell infiltration and immune checkpoint gene expression in both groups showed that both parameters were positively correlated with the risk level, indicating an increased immune response in higher risk groups. CONCLUSION Our findings suggest that microbial profiles in ovarian serous cystadenocarcinoma may serve as viable clinical prognostic indicators. This study provides novel insights into the potential impact of intratumoral microbial communities on disease prognosis and opens avenues for future therapeutic interventions targeting these microorganisms.
Collapse
Affiliation(s)
- Hao Qin
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| | - Jie Liu
- Department of Medical Records, Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Yi Qu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 North Huayuan Road, Haidian District, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology, (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yang-Yang Li
- Medical Center for Human Reproduction, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ya-Lan Xu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, China
| | - Yi-Fang Yan
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No.49 North Huayuan Road, Haidian District, Beijing, 100191, China.
- National Clinical Research Center for Obstetrics and Gynecology, (Peking University Third Hospital), Beijing, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
| |
Collapse
|
10
|
Xu J, Cheng M, Liu J, Cui M, Yin B, Liang J. Research progress on the impact of intratumoral microbiota on the immune microenvironment of malignant tumors and its role in immunotherapy. Front Immunol 2024; 15:1389446. [PMID: 39034996 PMCID: PMC11257860 DOI: 10.3389/fimmu.2024.1389446] [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: 02/22/2024] [Accepted: 06/21/2024] [Indexed: 07/23/2024] Open
Abstract
Microbiota has been closely related to human beings, whose role in tumor development has also been widely investigated. However, previous studies have mainly focused on the gut, oral, and/or skin microbiota. In recent years, the study of intratumoral microbiota has become a hot topic in tumor-concerning studies. Intratumoral microbiota plays an important role in the occurrence, development, and response to treatment of malignant tumors. In fact, increasing evidence has suggested that intratumoral microbiota is associated with malignant tumors in various ways, such as promoting the tumor development and affecting the efficacy of chemotherapy and immunotherapy. In this review, the impact of intratumoral microbiota on the immune microenvironment of malignant tumors has been analyzed, as well as its role in tumor immunotherapy, with the hope that it may contribute to the development of diagnostic tools and treatments for related tumors in the future.
Collapse
Affiliation(s)
- Jiamei Xu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, China
| | - Min Cheng
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, China
| | - Jie Liu
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Mengqi Cui
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, China
| | - Beibei Yin
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, China
| | - Jing Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, China
| |
Collapse
|
11
|
Glieca S, Quarta E, Bottari B, Lal VC, Sonvico F, Buttini F. The role of airways microbiota on local and systemic diseases: a rationale for probiotics delivery to the respiratory tract. Expert Opin Drug Deliv 2024; 21:991-1005. [PMID: 39041243 DOI: 10.1080/17425247.2024.2380334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Recent discoveries in the field of lung microbiota have enabled the investigation of new therapeutic interventions involving the use of inhaled probiotics. AREAS COVERED This review provides an overview of what is known about the correlation between airway dysbiosis and the development of local and systemic diseases, and how this knowledge can be exploited for therapeutic interventions. In particular, the review focused on attempts to formulate probiotics that can be deposited directly on the airways. EXPERT OPINION Despite considerable progress since the emergence of respiratory microbiota restoration as a new research field, numerous clinical implications and benefits remain to be determined. In the case of local diseases, once the pathophysiology is understood, manipulating the lung microbiota through probiotic administration is an approach that can be exploited. In contrast, the effect of pulmonary dysbiosis on systemic diseases remains to be clarified; however, this approach could represent a turning point in their treatment.
Collapse
Affiliation(s)
| | - Eride Quarta
- Food and Drug Department, University of Parma, Parma, Italy
| | | | | | - Fabio Sonvico
- Food and Drug Department, University of Parma, Parma, Italy
- Interdepartmental Center for Innovation in Health Products, Biopharmanet_TEC, University of Parma, Parma, Italy
| | - Francesca Buttini
- Food and Drug Department, University of Parma, Parma, Italy
- Interdepartmental Center for Innovation in Health Products, Biopharmanet_TEC, University of Parma, Parma, Italy
| |
Collapse
|
12
|
Yalamarty R, Magesh S, John D, Chakladar J, Li WT, Brumund KT, Wang-Rodriguez J, Ongkeko WM. The intratumor microbiome varies by geographical location and anatomical site in head and neck squamous cell carcinoma. Curr Probl Cancer 2024; 50:101100. [PMID: 38820649 DOI: 10.1016/j.currproblcancer.2024.101100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/29/2024] [Accepted: 04/25/2024] [Indexed: 06/02/2024]
Abstract
Head and Neck Squamous Cell Carcinoma (HNSCC) is a highly heterogeneous cancer that is characterized by distinct phenotypes based on anatomical site and etiological agents. Recently, the intratumor microbiome has been implicated in cancer pathogenesis and progression. Although it is well established that the gut microbiome varies with geographical location and is highly influenced by factors such as diet, environment, and genetics, the intratumor microbiome is not very well characterized. In this review, we aim to characterize the HNSCC intratumor microbiome by geographical location and anatomical site. We conducted a review of primary literature from PubMed and assessed studies based on relevancy and recency. To the best of our knowledge, we are the first to comprehensively examine the tumor microenvironment of HNSCC with respect to these two primary factors on a large scale. Our results suggest that there are unique bacterial and fungal biomarkers for HNSCC for each of the following geographical locations: North America, Asia, Europe, Australia, and Africa. We also identified a panel of microbial biomarkers that are unique to two primary HNSCC anatomic sites, as well as microbial biomarkers associated with various etiological agents of HNSCC. Future study of these microbes may improve HNSCC diagnostic and therapeutic modalities by accounting for differences based on geographic regions and anatomical sites.
Collapse
Affiliation(s)
- Rishabh Yalamarty
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of California, San Diego, CA 92093, USA; Research Service, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Shruti Magesh
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of California, San Diego, CA 92093, USA; Research Service, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Daniel John
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of California, San Diego, CA 92093, USA; Research Service, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Jaideep Chakladar
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of California, San Diego, CA 92093, USA; Research Service, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Wei Tse Li
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of California, San Diego, CA 92093, USA; Research Service, VA San Diego Healthcare System, San Diego, CA 92161, USA; University of California San Francisco School of Medicine, San Francisco, CA 94143, USA
| | - Kevin T Brumund
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of California, San Diego, CA 92093, USA; Division of Head and Neck Surgery, Department of Surgery, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Jessica Wang-Rodriguez
- Pathology Service, VA San Diego Healthcare System, San Diego, CA 92161, USA; Department of Pathology, UC San Diego School of Medicine, San Diego, CA 92093, USA
| | - Weg M Ongkeko
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of California, San Diego, CA 92093, USA; Research Service, VA San Diego Healthcare System, San Diego, CA 92161, USA.
| |
Collapse
|
13
|
Leoni C, Vinci L, Marzano M, D’Erchia AM, Dellino M, Cox SN, Vitagliano A, Visci G, Notario E, Filomena E, Cicinelli E, Pesole G, Ceci LR. Endometrial Cancer: A Pilot Study of the Tissue Microbiota. Microorganisms 2024; 12:1090. [PMID: 38930472 PMCID: PMC11205883 DOI: 10.3390/microorganisms12061090] [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: 04/26/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND The endometrium remains a difficult tissue for the analysis of microbiota, mainly due to the low bacterial presence and the sampling procedures. Among its pathologies, endometrial cancer has not yet been completely investigated for its relationship with microbiota composition. In this work, we report on possible correlations between endometrial microbiota dysbiosis and endometrial cancer. METHODS Women with endometrial cancer at various stages of tumor progression were enrolled together with women with a benign polymyomatous uterus as the control. Analyses were performed using biopsies collected at two specific endometrial sites during the surgery. This study adopted two approaches: the absolute quantification of the bacterial load, using droplet digital PCR (ddPCR), and the analysis of the bacterial composition, using a deep metabarcoding NGS procedure. RESULTS ddPCR provided the first-ever assessment of the absolute quantification of bacterial DNA in the endometrium, confirming a generally low microbial abundance. Metabarcoding analysis revealed a different microbiota distribution in the two endometrial sites, regardless of pathology, accompanied by an overall higher prevalence of pathogenic bacterial genera in cancerous tissues. CONCLUSIONS These results pave the way for future studies aimed at identifying potential biomarkers and gaining a deeper understanding of the role of bacteria associated with tumors.
Collapse
Affiliation(s)
- Claudia Leoni
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council (CNR), Via Amendola n. 122/O, 70126 Bari, Italy; (M.M.); (E.N.)
| | - Lorenzo Vinci
- 2nd Unit of Obstetrics and Gynaecology, Department of Biomedical Science and Human Oncology, University of Bari “A. Moro”, Piazza G. Cesare, 70124 Bari, Italy; (L.V.); (M.D.); (A.V.); (E.C.)
| | - Marinella Marzano
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council (CNR), Via Amendola n. 122/O, 70126 Bari, Italy; (M.M.); (E.N.)
| | - Anna Maria D’Erchia
- Department of Biosciences, Biotechnologies and Environment, University of Bari A. Moro, Via Orabona n. 4, 70126 Bari, Italy; (A.M.D.); (S.N.C.); (G.V.); (E.F.); (G.P.)
| | - Miriam Dellino
- 2nd Unit of Obstetrics and Gynaecology, Department of Biomedical Science and Human Oncology, University of Bari “A. Moro”, Piazza G. Cesare, 70124 Bari, Italy; (L.V.); (M.D.); (A.V.); (E.C.)
| | - Sharon Natasha Cox
- Department of Biosciences, Biotechnologies and Environment, University of Bari A. Moro, Via Orabona n. 4, 70126 Bari, Italy; (A.M.D.); (S.N.C.); (G.V.); (E.F.); (G.P.)
| | - Amerigo Vitagliano
- 2nd Unit of Obstetrics and Gynaecology, Department of Biomedical Science and Human Oncology, University of Bari “A. Moro”, Piazza G. Cesare, 70124 Bari, Italy; (L.V.); (M.D.); (A.V.); (E.C.)
| | - Grazia Visci
- Department of Biosciences, Biotechnologies and Environment, University of Bari A. Moro, Via Orabona n. 4, 70126 Bari, Italy; (A.M.D.); (S.N.C.); (G.V.); (E.F.); (G.P.)
| | - Elisabetta Notario
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council (CNR), Via Amendola n. 122/O, 70126 Bari, Italy; (M.M.); (E.N.)
| | - Ermes Filomena
- Department of Biosciences, Biotechnologies and Environment, University of Bari A. Moro, Via Orabona n. 4, 70126 Bari, Italy; (A.M.D.); (S.N.C.); (G.V.); (E.F.); (G.P.)
| | - Ettore Cicinelli
- 2nd Unit of Obstetrics and Gynaecology, Department of Biomedical Science and Human Oncology, University of Bari “A. Moro”, Piazza G. Cesare, 70124 Bari, Italy; (L.V.); (M.D.); (A.V.); (E.C.)
| | - Graziano Pesole
- Department of Biosciences, Biotechnologies and Environment, University of Bari A. Moro, Via Orabona n. 4, 70126 Bari, Italy; (A.M.D.); (S.N.C.); (G.V.); (E.F.); (G.P.)
| | - Luigi Ruggiero Ceci
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council (CNR), Via Amendola n. 122/O, 70126 Bari, Italy; (M.M.); (E.N.)
| |
Collapse
|
14
|
Wu K, Li Y, Ma K, Zhao W, Yao Z, Zheng Z, Sun F, Mu X, Liu Z, Zheng J. The microbiota and renal cell carcinoma. Cell Oncol (Dordr) 2024; 47:397-413. [PMID: 37878209 DOI: 10.1007/s13402-023-00876-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2023] [Indexed: 10/26/2023] Open
Abstract
Renal cell carcinoma (RCC) accounts for about 2% of cancer diagnoses and deaths worldwide. Recent studies emphasized the critical involvement of microbial populations in RCC from oncogenesis, tumor growth, and response to anticancer therapy. Microorganisms have been shown to be involved in various renal physiological and pathological processes by influencing the immune system function, metabolism of the host and pharmaceutical reactions. These findings have extended our understanding and provided more possibilities for the diagnostic or therapeutic development of microbiota, which could function as screening, prognostic, and predictive biomarkers, or be manipulated to prevent RCC progression, boost anticancer drug efficacy and lessen the side effects of therapy. This review aims to present an overview of the roles of microbiota in RCC, including pertinent mechanisms in microbiota-related carcinogenesis, the potential use of the microbiota as RCC biomarkers, and the possibility of modifying the microbiota for RCC prevention or treatment. According to these scientific findings, the clinical translation of microbiota is expected to improve the diagnosis and treatment of RCC.
Collapse
Affiliation(s)
- Ke Wu
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaorong Li
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kangli Ma
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiguang Zhao
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhixian Yao
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhong Zheng
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Sun
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingyu Mu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhihong Liu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Junhua Zheng
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
15
|
Liu J, Qu Y, Li YY, Xu YL, Yan YF, Qin H. Exploring prognostic microbiota markers in patients with endometrial carcinoma: Intratumoral insights. Heliyon 2024; 10:e27879. [PMID: 38515713 PMCID: PMC10955307 DOI: 10.1016/j.heliyon.2024.e27879] [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: 10/07/2023] [Revised: 03/03/2024] [Accepted: 03/07/2024] [Indexed: 03/23/2024] Open
Abstract
Endometrial cancer, a leading gynecological malignancy, is profoundly influenced by the uterine microbiota, a key factor in disease prognosis and treatment. Our study underscores the distinct microbial compositions in endometrial cancer compared to adjacent non-cancerous tissues, revealing a dominant presence of p_Actinobacteria in cancerous tissues as opposed to p_Firmicutes in surrounding areas. Through comprehensive analysis, we identified 485 unique microorganisms in cancer tissues, 26 of which correlate with patient prognosis. Employing univariate Cox regression and LASSO regression analyses, we devised a microbial risk scoring model, effectively stratifying patients into high and low-risk categories, thereby providing predictive insights into their overall survival. We further developed a nomogram that incorporates the microbial risk score along with age, grade, and clinical stage, significantly enhancing the accuracy of our clinical prediction model for endometrial cancer. Moreover, our study delves into the differential immune landscapes of high-risk and low-risk patients. The low-risk group displayed a higher prevalence of activated B cells and increased T cell co-stimulation, indicative of a robust immune response. Conversely, high-risk patients showed elevated tumor immune dysfunction and exclusion scores, suggesting less favorable outcomes in immunotherapy. Notably, the efficacy of IPS-CTLA4 and PD1/PD-L1/PD-L2 blockers was substantially higher in the low-risk group, pointing to a more responsive immunotherapeutic approach. In summary, our research elucidates the unique microbial patterns in endometrial cancer and adjacent tissues, and establishes both a microbial risk score model and a clinical prediction nomogram. These findings highlight the potential of uterine microbiota as a biomarker for customizing treatment strategies, enabling precise interventions for high-risk patients while preventing overtreatment in low-risk cases. This study emphasizes the microbiota's role in tailoring immunotherapy, offering a novel perspective in the treatment and prognosis of endometrial cancer. Significantly, our study's expansive sample analysis from the TCGA-UCEC cohort, employing linear discriminant analysis effect size methodology, not only validates but also enhances our understanding of the microbiota's role in endometrial cancer, paving the way for novel diagnostic and therapeutic approaches in its management.
Collapse
Affiliation(s)
- Jie Liu
- Department of Medical Records, Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Yi Qu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Yang-Yang Li
- Medical Center for Human Reproduction, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ya-Lan Xu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, China
| | - Yi-Fang Yan
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Hao Qin
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
Wu J, Zhang P, Mei W, Zeng C. Intratumoral microbiota: implications for cancer onset, progression, and therapy. Front Immunol 2024; 14:1301506. [PMID: 38292482 PMCID: PMC10824977 DOI: 10.3389/fimmu.2023.1301506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/28/2023] [Indexed: 02/01/2024] Open
Abstract
Significant advancements have been made in comprehending the interactions between the microbiome and cancer. However, prevailing research predominantly directs its focus toward the gut microbiome, affording limited consideration to the interactions of intratumoral microbiota and tumors. Within the tumor microenvironment (TME), the intratumoral microbiome and its associated products wield regulatory influence, directing the modulation of cancer cell properties and impacting immune system functionality. However, to grasp a more profound insight into the intratumoral microbiota in cancer, further research into its underlying mechanisms is necessary. In this review, we delve into the intricate associations between intratumoral microbiota and cancer, with a specific focus on elucidating the significant contribution of intratumoral microbiota to the onset and advancement of cancer. Notably, we provide a detailed exploration of therapeutic advances facilitated by intratumoral microbiota, offering insights into recent developments in this burgeoning field.
Collapse
Affiliation(s)
- Jinmei Wu
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Pengfei Zhang
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Wuxuan Mei
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen, China
| |
Collapse
|
17
|
Sevcikova A, Mladosievicova B, Mego M, Ciernikova S. Exploring the Role of the Gut and Intratumoral Microbiomes in Tumor Progression and Metastasis. Int J Mol Sci 2023; 24:17199. [PMID: 38139030 PMCID: PMC10742837 DOI: 10.3390/ijms242417199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/30/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Cancer cell dissemination involves invasion, migration, resistance to stressors in the circulation, extravasation, colonization, and other functions responsible for macroscopic metastases. By enhancing invasiveness, motility, and intravasation, the epithelial-to-mesenchymal transition (EMT) process promotes the generation of circulating tumor cells and their collective migration. Preclinical and clinical studies have documented intensive crosstalk between the gut microbiome, host organism, and immune system. According to the findings, polymorphic microbes might play diverse roles in tumorigenesis, cancer progression, and therapy response. Microbial imbalances and changes in the levels of bacterial metabolites and toxins promote cancer progression via EMT and angiogenesis. In contrast, a favorable microbial composition, together with microbiota-derived metabolites, such as short-chain fatty acids (SCFAs), can attenuate the processes of tumor initiation, disease progression, and the formation of distant metastases. In this review, we highlight the role of the intratumoral and gut microbiomes in cancer cell invasion, migration, and metastatic ability and outline the potential options for microbiota modulation. As shown in murine models, probiotics inhibited tumor development, reduced tumor volume, and suppressed angiogenesis and metastasis. Moreover, modulation of an unfavorable microbiome might improve efficacy and reduce treatment-related toxicities, bringing clinical benefit to patients with metastatic cancer.
Collapse
Affiliation(s)
- Aneta Sevcikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia;
| | - Beata Mladosievicova
- Institute of Pathological Physiology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia;
| | - Michal Mego
- 2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia;
| | - Sona Ciernikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia;
| |
Collapse
|
18
|
Meng YF, Fan ZY, Zhou B, Zhan HX. Role of the intratumoral microbiome in tumor progression and therapeutics implications. Biochim Biophys Acta Rev Cancer 2023; 1878:189014. [PMID: 37918451 DOI: 10.1016/j.bbcan.2023.189014] [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: 06/05/2023] [Revised: 10/20/2023] [Accepted: 10/20/2023] [Indexed: 11/04/2023]
Abstract
Microbes are widely present in various organs of the human body and play important roles in numerous physiological and pathological processes. Nevertheless, owing to multiple limiting factors, such as contamination and low biomass, the current understanding of the intratumoral microbiome is limited. The intratumoral microbiome exerts tumor-promoting or tumor-suppressive effects by engaging in metabolic reactions within the body, regulating signaling cancer-related pathways, and impacting both host cells function and immune system. It is important to emphasize that intratumoral microbes exhibit substantial heterogeneity in terms of composition and abundance across various tumor types, thereby potentially influencing diverse aspects of tumorigenesis, progression, and metastasis. These findings suggest that intratumoral microbiome have great potential as diagnostic and prognostic biomarkers. By manipulating the intratumoral microbes to employ cancer therapy, the efficacy of chemotherapy or immunotherapy can be enhanced while minimizing adverse effects. In this review, we comprehensively describe the composition and function of the intratumoral microbiome in various human solid tumors. Combining recent advancements in research, we discuss the origins, mechanisms, and prospects of the clinical applications of intratumoral microbiome.
Collapse
Affiliation(s)
- Yu-Fan Meng
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Zhi-Yao Fan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Department of Retroperitoneal Tumor Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Han-Xiang Zhan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
19
|
Zhu J, Liu W, Bian Z, Ma Y, Kang Z, Jin J, Li X, Ge S, Hao Y, Zhang H, Xie Y. Lactobacillus plantarum Zhang-LL Inhibits Colitis-Related Tumorigenesis by Regulating Arachidonic Acid Metabolism and CD22-Mediated B-Cell Receptor Regulation. Nutrients 2023; 15:4512. [PMID: 37960165 PMCID: PMC10648432 DOI: 10.3390/nu15214512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/07/2023] [Accepted: 10/11/2023] [Indexed: 11/15/2023] Open
Abstract
Colorectal cancer (CRC) is a significant health concern and is the third most commonly diagnosed and second deadliest cancer worldwide. CRC has been steadily increasing in developing countries owing to factors such as aging and epidemics. Despite extensive research, the exact pathogenesis of CRC remains unclear, and its causes are complex and variable. Numerous in vitro, animal, and clinical trials have demonstrated the efficacy of probiotics such as Lactobacillus plantarum in reversing the adverse outcomes of CRC. These findings suggest that probiotics play vital roles in the prevention, adjuvant treatment, and prognosis of CRC. In this study, we constructed a mouse model of CRC using an intraperitoneal injection of azomethane combined with dextran sodium sulfate, while administering 5-fluorouracil as well as high- and low-doses of L. plantarum Zhang-LL live or heat-killed strains. Weight changes and disease activity indices were recorded during feeding, and the number of polyps and colon length were measured after euthanasia. HE staining was used to observe the histopathological changes in the colons of mice, and ELISA was used to detect the expression levels of IL-1β, TNF-α, and IFN-γ in serum. To investigate the specific mechanisms involved in alleviating CRC progression, gut microbial alterations were investigated using 16S rRNA amplicon sequencing and non-targeted metabolomics, and changes in genes related to CRC were assessed using eukaryotic transcriptomics. The results showed that both viable and heat-killed strains of L. plantarum Zhang-LL in high doses significantly inhibited tumorigenesis, colon shortening, adverse inflammatory reactions, intestinal tissue damage, and pro-inflammatory factor expression upregulation. Specifically, in the gut microbiota, the abundance of the dominant flora Acutalibacter muris and Lactobacillus johnsonii was regulated, PGE2 expression was significantly reduced, the arachidonic acid metabolism pathway was inhibited, and CD22-mediated B-cell receptor regulation-related gene expression was upregulated. This study showed that L. plantarum Zhang-LL live or heat-inactivated strains alleviated CRC progression by reducing the abundance of potentially pathogenic bacteria, increasing the abundance of beneficial commensal bacteria, mediating the arachidonic acid metabolism pathway, and improving host immunogenicity.
Collapse
Affiliation(s)
- Jingxin Zhu
- Beijing Laboratory of Food Quality and Safety, Beijing Key Laboratory of Agricultural Product Detection and Control of Spoilage Organisms and Pesticide Residue, College of Food Science and Engineering, Beijing University of Agriculture, Beijing 102206, China; (J.Z.); (W.L.); (Z.B.); (Y.M.); (Z.K.); (J.J.); (X.L.)
| | - Wenbo Liu
- Beijing Laboratory of Food Quality and Safety, Beijing Key Laboratory of Agricultural Product Detection and Control of Spoilage Organisms and Pesticide Residue, College of Food Science and Engineering, Beijing University of Agriculture, Beijing 102206, China; (J.Z.); (W.L.); (Z.B.); (Y.M.); (Z.K.); (J.J.); (X.L.)
| | - Zheng Bian
- Beijing Laboratory of Food Quality and Safety, Beijing Key Laboratory of Agricultural Product Detection and Control of Spoilage Organisms and Pesticide Residue, College of Food Science and Engineering, Beijing University of Agriculture, Beijing 102206, China; (J.Z.); (W.L.); (Z.B.); (Y.M.); (Z.K.); (J.J.); (X.L.)
| | - Yumeng Ma
- Beijing Laboratory of Food Quality and Safety, Beijing Key Laboratory of Agricultural Product Detection and Control of Spoilage Organisms and Pesticide Residue, College of Food Science and Engineering, Beijing University of Agriculture, Beijing 102206, China; (J.Z.); (W.L.); (Z.B.); (Y.M.); (Z.K.); (J.J.); (X.L.)
| | - Zixin Kang
- Beijing Laboratory of Food Quality and Safety, Beijing Key Laboratory of Agricultural Product Detection and Control of Spoilage Organisms and Pesticide Residue, College of Food Science and Engineering, Beijing University of Agriculture, Beijing 102206, China; (J.Z.); (W.L.); (Z.B.); (Y.M.); (Z.K.); (J.J.); (X.L.)
| | - Junhua Jin
- Beijing Laboratory of Food Quality and Safety, Beijing Key Laboratory of Agricultural Product Detection and Control of Spoilage Organisms and Pesticide Residue, College of Food Science and Engineering, Beijing University of Agriculture, Beijing 102206, China; (J.Z.); (W.L.); (Z.B.); (Y.M.); (Z.K.); (J.J.); (X.L.)
| | - Xiangyang Li
- Beijing Laboratory of Food Quality and Safety, Beijing Key Laboratory of Agricultural Product Detection and Control of Spoilage Organisms and Pesticide Residue, College of Food Science and Engineering, Beijing University of Agriculture, Beijing 102206, China; (J.Z.); (W.L.); (Z.B.); (Y.M.); (Z.K.); (J.J.); (X.L.)
| | - Shaoyang Ge
- Beijing HEYIYUAN BIOTECHNOLOGY Co., Ltd., Beijing 100088, China;
| | - Yanling Hao
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, Co-Constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing 100190, China;
| | - Hongxing Zhang
- Beijing Laboratory of Food Quality and Safety, Beijing Key Laboratory of Agricultural Product Detection and Control of Spoilage Organisms and Pesticide Residue, College of Food Science and Engineering, Beijing University of Agriculture, Beijing 102206, China; (J.Z.); (W.L.); (Z.B.); (Y.M.); (Z.K.); (J.J.); (X.L.)
| | - Yuanhong Xie
- Beijing Laboratory of Food Quality and Safety, Beijing Key Laboratory of Agricultural Product Detection and Control of Spoilage Organisms and Pesticide Residue, College of Food Science and Engineering, Beijing University of Agriculture, Beijing 102206, China; (J.Z.); (W.L.); (Z.B.); (Y.M.); (Z.K.); (J.J.); (X.L.)
| |
Collapse
|
20
|
Ji H, Jiang Z, Wei C, Ma Y, Zhao J, Wang F, Zhao B, Wang D, Tang D. Intratumoural microbiota: from theory to clinical application. Cell Commun Signal 2023; 21:164. [PMID: 37381018 DOI: 10.1186/s12964-023-01134-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/22/2023] [Indexed: 06/30/2023] Open
Abstract
Cancer is a major cause of high morbidity and mortality worldwide. Several environmental, genetic and lifestyle factors are associated with the development of cancer in humans and result in suboptimal treatment. The human microbiota has been implicated in the pathophysiological process of cancer and has been used as a diagnostic, prognostic and risk assessment tool in cancer management. Notably, both extratumoural and intratumoural microbiota are important components of the tumor microenvironment, subtly influencing tumorigenesis, progression, treatment and prognosis. The potential oncogenic mechanisms of action of the intratumoural microbiota include induction of DNA damage, influence on cell signaling pathways and impairment of immune responses. Some naturally occurring or genetically engineered microorganisms can specifically accumulate and replicate in tumors and then initiate various anti-tumor programs, ultimately promoting the therapeutic effect of tumor microbiota and reducing the toxic and side effects of conventional tumor treatments, which may be conducive to the pursuit of accurate cancer treatment. In this review, we summarise evidence revealing the impact of the intratumoural microbiota on cancer occurrence and progress and potential therapeutic and diagnostic applications, which may be a promising novel strategy to inhibit tumor development and enhance therapeutic efficacy. Video Abstract.
Collapse
Affiliation(s)
- Hao Ji
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu Province, China
| | - Zhengting Jiang
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu Province, China
| | - Chen Wei
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu Province, China
| | - Yichao Ma
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu Province, China
| | - Jiahao Zhao
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu Province, China
| | - Fei Wang
- Clinical Medical College, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Bin Zhao
- Clinical Medical College, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225000, China.
| |
Collapse
|
21
|
Bhoopathi P, Mannangatti P, Das SK, Fisher PB, Emdad L. Chemoresistance in pancreatic ductal adenocarcinoma: Overcoming resistance to therapy. Adv Cancer Res 2023; 159:285-341. [PMID: 37268399 DOI: 10.1016/bs.acr.2023.02.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a prominent cause of cancer deaths worldwide, is a highly aggressive cancer most frequently detected at an advanced stage that limits treatment options to systemic chemotherapy, which has provided only marginal positive clinical outcomes. More than 90% of patients with PDAC die within a year of being diagnosed. PDAC is increasing at a rate of 0.5-1.0% per year, and it is expected to be the second leading cause of cancer-related mortality by 2030. The resistance of tumor cells to chemotherapeutic drugs, which can be innate or acquired, is the primary factor contributing to the ineffectiveness of cancer treatments. Although many PDAC patients initially responds to standard of care (SOC) drugs they soon develop resistance caused partly by the substantial cellular heterogeneity seen in PDAC tissue and the tumor microenvironment (TME), which are considered key factors contributing to resistance to therapy. A deeper understanding of molecular mechanisms involved in PDAC progression and metastasis development, and the interplay of the TME in all these processes is essential to better comprehend the etiology and pathobiology of chemoresistance observed in PDAC. Recent research has recognized new therapeutic targets ushering in the development of innovative combinatorial therapies as well as enhancing our comprehension of several different cell death pathways. These approaches facilitate the lowering of the therapeutic threshold; however, the possibility of subsequent resistance development still remains a key issue and concern. Discoveries, that can target PDAC resistance, either alone or in combination, have the potential to serve as the foundation for future treatments that are effective without posing undue health risks. In this chapter, we discuss potential causes of PDAC chemoresistance and approaches for combating chemoresistance by targeting different pathways and different cellular functions associated with and mediating resistance.
Collapse
Affiliation(s)
- Praveen Bhoopathi
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States
| | - Padmanabhan Mannangatti
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
22
|
Chen F, Yang J, Guo Y, Su D, Sheng Y, Wu Y. Integrating bulk and single-cell RNA sequencing data reveals the relationship between intratumor microbiome signature and host metabolic heterogeneity in breast cancer. Front Immunol 2023; 14:1140995. [PMID: 36999009 PMCID: PMC10049788 DOI: 10.3389/fimmu.2023.1140995] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
IntroductionNowadays, it has been recognized that gut microbiome can indirectly modulate cancer susceptibility or progression. However, whether intratumor microbes are parasitic, symbiotic, or merely bystanders in breast cancer is not fully understood. Microbial metabolite plays a pivotal role in the interaction of host and microbe via regulating mitochondrial and other metabolic pathways. And the relationship between tumor-resident microbiota and cancer metabolism remains an open question.Methods1085 breast cancer patients with normalized intratumor microbial abundance data and 32 single-cell RNA sequencing samples were retrieved from public datasets. We used the gene set variation analysis to evaluate the various metabolic activities of breast cancer samples. Furthermore, we applied Scissor method to identify microbe-associated cell subpopulations from single-cell data. Then, we conducted comprehensive bioinformatic analyses to explore the association between host and microbe in breast cancer.ResultsHere, we found that the metabolic status of breast cancer cells was highly plastic, and some microbial genera were significantly correlated with cancer metabolic activity. We identified two distinct clusters based on microbial abundance and tumor metabolism data. And dysregulation of the metabolic pathway was observed among different cell types. Metabolism-related microbial scores were calculated to predict overall survival in patients with breast cancer. Furthermore, the microbial abundance of the specific genus was associated with gene mutation due to possible microbe-mediated mutagenesis. The infiltrating immune cell compositions, including regulatory T cells and activated NK cells, were significantly associated with the metabolism-related intratumor microbes, as indicated in the Mantel test analysis. Moreover, the mammary metabolism-related microbes were related to T cell exclusion and response to immunotherapy.ConclusionsOverall, the exploratory study shed light on the potential role of the metabolism-related microbiome in breast cancer patients. And the novel treatment will be realized by further investigating the metabolic disturbance in host and intratumor microbial cells.
Collapse
Affiliation(s)
- Fangyue Chen
- Department of General Surgery, Changhai Hospital, Navy Military Medical University, Shanghai, China
| | - Jun Yang
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Youxiang Guo
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Dongwei Su
- Department of General Surgery, Changhai Hospital, Navy Military Medical University, Shanghai, China
| | - Yuan Sheng
- Department of General Surgery, Changhai Hospital, Navy Military Medical University, Shanghai, China
- *Correspondence: Yuan Sheng, ; Yanmei Wu,
| | - Yanmei Wu
- Department of General Surgery, Changhai Hospital, Navy Military Medical University, Shanghai, China
- *Correspondence: Yuan Sheng, ; Yanmei Wu,
| |
Collapse
|