1
|
Preet R, Islam MA, Shim J, Rajendran G, Mitra A, Vishwakarma V, Kutz C, Choudhury S, Pathak H, Dai Q, Sun W, Madan R, Zhong C, Markiewicz MA, Zhang J. Gut commensal Bifidobacterium-derived extracellular vesicles modulate the therapeutic effects of anti-PD-1 in lung cancer. Nat Commun 2025; 16:3500. [PMID: 40221398 PMCID: PMC11993705 DOI: 10.1038/s41467-025-58553-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Although immunotherapy such as anti-programmed death-1 and its ligand 1 (PD-1/L1) is a standard treatment for advanced non-small cell lung cancer (NSCLC), many patients do not derive benefit directly. Several studies have elucidated new strategies to improve the antitumor immune response through gut microbiota modulation. However, it remains largely debatable regarding how gut microbiota remotely affect lung cancer microenvironment and subsequently modulate immunotherapy response. Here we show that commensal Bifidobacterium-derived extracellular vesicles (Bif.BEVs) can modulate the therapeutic effect of anti-PD-1 therapy in NSCLC. These Bif.BEVs are up-taken by lung cancer cells predominantly via dynamin-dependent endocytosis and upregulate PD-L1 expression through TLR4-NF-κB pathway. They also efficiently penetrate murine intestinal and patient-derived lung cancer organoids. Oral gavage of these Bif.BEVs result in their accumulation in tumors in mice. Using a syngeneic mouse model, Bif.BEVs are found to synergize the anti-tumor effect of anti-PD-1 via modulation of key cytokines, immune response and oncogenic pathways, and increase in tumor-infiltrating CD8+ T cells. Our study therefore identifies a link between Bif.BEVs and the tumor microenvironment, providing an alternative mechanism to explain how gut microbiota can influence immunotherapy response, particularly in tumors located anatomically distant from the gut.
Collapse
Affiliation(s)
- Ranjan Preet
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Md Atiqul Islam
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Jiyoung Shim
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Ganeshkumar Rajendran
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Amrita Mitra
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Vikalp Vishwakarma
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Caleb Kutz
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Sonali Choudhury
- Department of Cancer Biology, University of Kansas Comprehensive Cancer Center, Kansas City, KS, 66160, USA
| | - Harsh Pathak
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Qun Dai
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Weijing Sun
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Rashna Madan
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Cuncong Zhong
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, 66045, USA
| | - Mary A Markiewicz
- Department of Microbiology, Molecular Genetics & Immunology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Jun Zhang
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
- Department of Cancer Biology, University of Kansas Comprehensive Cancer Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
2
|
Wang S, Tan X, Cheng J, Liu Z, Zhou H, Liao J, Wang X, Liu H. Oral microbiome and its relationship with oral cancer. J Cancer Res Ther 2024; 20:1141-1149. [PMID: 39206975 DOI: 10.4103/jcrt.jcrt_44_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 07/01/2024] [Indexed: 09/04/2024]
Abstract
ABSTRACT As the initial point for digestion, the balance of oral microorganisms plays an important role in maintaining local and systemic health. Oral dysbiosis, or an imbalance in the oral microbial community, may lead to the onset of various diseases. The presence or abnormal increase of microbes in the oral cavity has attracted significant attention due to its complicated relationship with oral cancer. Oral cancer can remodel microbial profiles by creating a more beneficial microenvironment for its progression. On the other hand, altered microbial profiles can promote tumorigenesis by evoking a complex inflammatory response and affecting host immunity. This review analyzes the oncogenic potential of oral microbiome alterations as a driver and biomarker. Additionally, a potentially therapeutic strategy via the reversal of the oral microbiome dysbiosis in oral cancers has been discussed.
Collapse
Affiliation(s)
- Shengran Wang
- School of Stomatology and Ophthalmology, Xianning Medical College, Hubei University of Science and Technology, Xianning Hubei, China
| | - Xiao Tan
- School of Clinical Medicine, Xianning Medical College, Hubei University of Science and Technology, Xianning Hubei, China
| | - Juan Cheng
- School of Clinical Medicine, Xianning Medical College, Hubei University of Science and Technology, Xianning Hubei, China
| | - Zeyang Liu
- School of Clinical Medicine, Xianning Medical College, Hubei University of Science and Technology, Xianning Hubei, China
| | - Huiping Zhou
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning Hubei, China
| | - Jiyuan Liao
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning Hubei, China
| | - Xijun Wang
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning Hubei, China
| | - Hongyun Liu
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning Hubei, China
| |
Collapse
|
3
|
Syed M, Cagely M, Dogra P, Hollmer L, Butner JD, Cristini V, Koay EJ. Immune-checkpoint inhibitor therapy response evaluation using oncophysics-based mathematical models. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1855. [PMID: 36148978 PMCID: PMC11824897 DOI: 10.1002/wnan.1855] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 06/10/2022] [Accepted: 08/23/2022] [Indexed: 11/08/2022]
Abstract
The field of oncology has transformed with the advent of immunotherapies. The standard of care for multiple cancers now includes novel drugs that target key checkpoints that function to modulate immune responses, enabling the patient's immune system to elicit an effective anti-tumor response. While these immune-based approaches can have dramatic effects in terms of significantly reducing tumor burden and prolonging survival for patients, the therapeutic approach remains active only in a minority of patients and is often not durable. Multiple biological investigations have identified key markers that predict response to the most common form of immunotherapy-immune checkpoint inhibitors (ICI). These biomarkers help enrich patients for ICI but are not 100% predictive. Understanding the complex interactions of these biomarkers with other pathways and factors that lead to ICI resistance remains a major goal. Principles of oncophysics-the idea that cancer can be described as a multiscale physical aberration-have shown promise in recent years in terms of capturing the essence of the complexities of ICI interactions. Here, we review the biological knowledge of mechanisms of ICI action and how these are incorporated into modern oncophysics-based mathematical models. Building on the success of oncophysics-based mathematical models may help to discover new, rational methods to engineer immunotherapy for patients in the future. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Mustafa Syed
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Matthew Cagely
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, USA
| | - Lauren Hollmer
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Joseph D. Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, New York, USA
| | - Eugene J. Koay
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
4
|
Liu B, Chau J, Dai Q, Zhong C, Zhang J. Exploring Gut Microbiome in Predicting the Efficacy of Immunotherapy in Non-Small Cell Lung Cancer. Cancers (Basel) 2022; 14:5401. [PMID: 36358819 PMCID: PMC9656313 DOI: 10.3390/cancers14215401] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/29/2022] [Accepted: 10/29/2022] [Indexed: 11/02/2023] Open
Abstract
We performed various analyses on the taxonomic and functional features of the gut microbiome from NSCLC patients treated with immunotherapy to establish a model that may predict whether a patient will benefit from immunotherapy. We collected 65 published whole metagenome shotgun sequencing samples along with 14 samples from our previous study. We systematically studied the taxonomical characteristics of the dataset and used both the random forest (RF) and the multilayer perceptron (MLP) neural network models to predict patients with progression-free survival (PFS) above 6 months versus those below 3 months. Our results showed that the RF classifier achieved the highest F-score (85.2%) and the area under the receiver operating characteristic curve (AUC) (95%) using the protein families (Pfam) profile, and the MLP neural network classifier achieved a 99.9% F-score and 100% AUC using the same Pfam profile. When applying the model trained in the Pfam profile directly to predict the treatment response, we found that both trained RF and MLP classifiers significantly outperformed the stochastic predictor in F-score. Our results suggested that such a predictive model based on functional (e.g., Pfam) rather than taxonomic profile might be clinically useful to predict whether an NSCLC patient will benefit from immunotherapy, as both the F-score and AUC of functional profile outperform that of taxonomic profile. In addition, our model suggested that interactive biological processes such as methanogenesis, one-carbon, and amino acid metabolism might be important in regulating the immunotherapy response that warrants further investigation.
Collapse
Affiliation(s)
- Ben Liu
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS 66045, USA
| | - Justin Chau
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Qun Dai
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Cuncong Zhong
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS 66045, USA
- Bioengineering Program, School of Engineering, University of Kansas, Lawrence, KS 66045, USA
- Center for Computational Biology, University of Kansa, Lawrence, KS 66045, USA
| | - Jun Zhang
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
5
|
The Crosstalk between Microbiome and Immunotherapeutics: Myth or Reality. Cancers (Basel) 2022; 14:cancers14194641. [PMID: 36230563 PMCID: PMC9563484 DOI: 10.3390/cancers14194641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/21/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
The gut microbiome refers to microorganisms and their genetic material influencing local and systemic inflammation. Inflammation is known to contribute to cancer development, progression, and treatment. Evidence suggests that modulating the gut microbiome may affect responses to various cancer therapies. The gut microbiota has been suggested to have an impact on immunotherapy efficacy, especially the currently widely used immune checkpoint inhibitors in various malignancies. Microbial interventions like fecal microbiota transplantation, various probiotics, or even antibiotics can increase or decrease the tumor’s sensitivity to immunotherapy. However, not all tumors react in the same manner, highlighting the tumor microenvironment heterogeneity across tumor types and the influence this has on the crosstalk between the microbiome and therapy outcomes. In this study, we intend to review the association between the gut microbiota and immunotherapy response in cancer patients and the factors regulating this interaction.
Collapse
|
6
|
Wu H, Ganguly S, Tollefsbol TO. Modulating Microbiota as a New Strategy for Breast Cancer Prevention and Treatment. Microorganisms 2022; 10:microorganisms10091727. [PMID: 36144329 PMCID: PMC9503838 DOI: 10.3390/microorganisms10091727] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/18/2022] Open
Abstract
Breast cancer (BC) is the most common cancer in women in the United States. There has been an increasing incidence and decreasing mortality rate of BC cases over the past several decades. Many risk factors are associated with BC, such as diet, aging, personal and family history, obesity, and some environmental factors. Recent studies have shown that healthy individuals and BC patients have different microbiota composition, indicating that microbiome is a new risk factor for BC. Gut and breast microbiota alterations are associated with BC prognosis. This review will evaluate altered microbiota populations in gut, breast tissue, and milk of BC patients, as well as mechanisms of interactions between microbiota modulation and BC. Probiotics and prebiotics are commercially available dietary supplements to alleviate side-effects of cancer therapies. They also shape the population of human gut microbiome. This review evaluates novel means of modulating microbiota by nutritional treatment with probiotics and prebiotics as emerging and promising strategies for prevention and treatment of BC. The mechanistic role of probiotic and prebiotics partially depend on alterations in estrogen metabolism, systematic immune regulation, and epigenetics regulation.
Collapse
Affiliation(s)
- Huixin Wu
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA
| | - Sebanti Ganguly
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA
| | - Trygve O. Tollefsbol
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA
- Integrative Center for Aging Research, University of Alabama Birmingham, 1530 3rd Avenue South, Birmingham, AL 35294, USA
- O’Neal Comprehensive Cancer Center, University of Alabama Birmingham, 1802 6th Avenue South, Birmingham, AL 35294, USA
- Nutrition Obesity Research Center, University of Alabama Birmingham, 1675 University Boulevard, Birmingham, AL 35294, USA
- Comprehensive Diabetes Center, University of Alabama Birmingham, 1825 University Boulevard, Birmingham, AL 35294, USA
- University Wide Microbiome Center, University of Alabama Birmingham, 845 19th Street South, Birmingham, AL 35294, USA
- Correspondence: ; Tel.: +1-205-934-4573; Fax: +1-205-975-6097
| |
Collapse
|
7
|
Burcher KM, Burcher JT, Inscore L, Bloomer CH, Furdui CM, Porosnicu M. A Review of the Role of Oral Microbiome in the Development, Detection, and Management of Head and Neck Squamous Cell Cancers. Cancers (Basel) 2022; 14:4116. [PMID: 36077651 PMCID: PMC9454796 DOI: 10.3390/cancers14174116] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
The role of the microbiome in the development and propagation of head and neck squamous cell cancer (HNSCC) is largely unknown and the surrounding knowledge lags behind what has been discovered related to the microbiome and other malignancies. In this review, the authors performed a structured analysis of the available literature from several databases. The authors discuss the merits and detriments of several studies discussing the microbiome of the structures of the aerodigestive system throughout the development of HNSCC, the role of the microbiome in the development of malignancies (generally and in HNSCC) and clinical applications of the microbiome in HNSCC. Further studies will be needed to adequately describe the relationship between HNSCC and the microbiome, and to push this relationship into a space where it is clinically relevant outside of a research environment.
Collapse
Affiliation(s)
| | | | - Logan Inscore
- Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA
| | | | | | | |
Collapse
|
8
|
Song X, Wei C, Li X. The Relationship Between Microbial Community and Breast Cancer. Front Cell Infect Microbiol 2022; 12:849022. [PMID: 35782150 PMCID: PMC9245449 DOI: 10.3389/fcimb.2022.849022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/24/2022] [Indexed: 11/18/2022] Open
Abstract
Breast cancer (BC) is the most common cancer in women and the leading cause of cancer-related deaths in women worldwide. Recent research studies have shown that the intestinal flora is related to the occurrence and progression of BC. Notably, some evidence identifies a unique microbial community in breast tissue, a site previously thought to be sterile. In addition, breast tumors have their own specific microbial community, distinct from normal mammary gland tissue, and all of them may result from intestinal flora. Some microbial community in breast tissue may lead to the occurrence and development of BC. This review focuses on the relationship between the microbial community and breast cancer, which will lay a solid theoretical foundation for further understanding the local microenvironment of BC and developing effective targeted therapeutic drugs.
Collapse
Affiliation(s)
- Xuelian Song
- Department of The Graduate Student, Shandong First Medical University, Tai’an, China
| | - Changran Wei
- Department of The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiangqi Li
- Department of Breast Surgery, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| |
Collapse
|
9
|
Costa B, Vale N. Drug Metabolism for the Identification of Clinical Biomarkers in Breast Cancer. Int J Mol Sci 2022; 23:3181. [PMID: 35328602 PMCID: PMC8951384 DOI: 10.3390/ijms23063181] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/09/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Breast cancer is classified into four major molecular subtypes, and is considered a heterogenous disease. The risk profiles and treatment of breast cancer differ according to these subtypes. Early detection dramatically improves the prospects of successful treatment, resulting in a reduction in overall mortality rates. However, almost 30% of women primarily diagnosed with the early-stage disease will eventually develop metastasis or resistance to chemotherapies. Immunotherapies are among the most promising cancer treatment options; however, long-term clinical benefit has only been observed in a small subset of responding patients. The current strategies for diagnosis and treatment rely heavily on histopathological examination and molecular diagnosis, disregarding the tumor microenvironment and microbiome involving cancer cells. In this review, we aim to praise the use of pharmacogenomics and pharmacomicrobiomics as a strategy to identify potential biomarkers for guiding and monitoring therapy in real-time. The finding of these biomarkers can be performed by studying the metabolism of drugs, more specifically, immunometabolism, and its relationship with the microbiome, without neglecting the information provided by genetics. A larger understanding of cancer biology has the potential to improve patient care, enable clinical decisions, and deliver personalized medicine.
Collapse
Affiliation(s)
- Bárbara Costa
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal;
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal;
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- Associate Laboratory RISE-Health Research Network, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
10
|
Zong Y, Zhou Y, Liao B, Liao M, Shi Y, Wei Y, Huang Y, Zhou X, Cheng L, Ren B. The Interaction Between the Microbiome and Tumors. Front Cell Infect Microbiol 2021; 11:673724. [PMID: 34532297 PMCID: PMC8438519 DOI: 10.3389/fcimb.2021.673724] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 08/09/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer is a significant global health problem and is characterized by a consistent increase in incidence and mortality rate. Deciphering the etiology and risk factors are essential parts of cancer research. Recently, the altered microbiome has been identified within the tumor microenvironment, tumor tissue, and even nonadjacent environments, which indicates a strong correlation between the microbiome and tumor development. However, the causation and mechanisms of this correlation remain unclear. Herein, we summarized and discussed the interaction between the microbiome and tumor progression. Firstly, the microbiome, which can be located in the tumor microenvironment, inside tumor tissues and in the nonadjacent environment, is different between cancer patients and healthy individuals. Secondly, the tumor can remodel microbial profiles by creating a more beneficial condition for the shifted microbiome. Third, the microbiome can promote tumorigenesis through a direct pathogenic process, including the establishment of an inflammatory environment and its effect on host immunity. The interactions between the microbiome and tumors can promote an understanding of the carcinogenesis and provide novel therapeutic strategies for cancers.
Collapse
Affiliation(s)
- Yawen Zong
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Yujie Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Binyou Liao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Min Liao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Yangyang Shi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Yu Wei
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Yuyao Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Biao Ren
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Chau J, Zhang J. Tying Small Changes to Large Outcomes: The Cautious Promise in Incorporating the Microbiome into Immunotherapy. Int J Mol Sci 2021; 22:ijms22157900. [PMID: 34360663 PMCID: PMC8347117 DOI: 10.3390/ijms22157900] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 12/16/2022] Open
Abstract
The role of the microbiome in immunology is a rapidly burgeoning topic of study. Given the increasing use of immune checkpoint inhibitor (ICI) therapy in cancers, along with the recognition that carcinogenesis has been linked to dysregulations of the immune system, much attention is now directed at potentiation of ICI efficacy, as well as minimizing the incidence of treatment-associated immune-related adverse events (irAEs). We provide an overview of the major research establishing links between the microbiome to tumorigenesis, chemotherapy and radiation potentiation, and ICI efficacy and irAE development.
Collapse
Affiliation(s)
- Justin Chau
- Division of Hematology, Oncology and Blood & Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA 52246, USA;
| | - Jun Zhang
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Cancer Biology, University of Kansas Cancer Center, Kansas City, KS 66160, USA
- Correspondence: ; Tel.: +1-(913)-588-8150; Fax: +1-(913)-588-4085
| |
Collapse
|
12
|
Chau J, Yadav M, Liu B, Furqan M, Dai Q, Shahi S, Gupta A, Mercer KN, Eastman E, Hejleh TA, Chan C, Weiner GJ, Cherwin C, Lee STM, Zhong C, Mangalam A, Zhang J. Prospective correlation between the patient microbiome with response to and development of immune-mediated adverse effects to immunotherapy in lung cancer. BMC Cancer 2021; 21:808. [PMID: 34256732 PMCID: PMC8278634 DOI: 10.1186/s12885-021-08530-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/21/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Though the gut microbiome has been associated with efficacy of immunotherapy (ICI) in certain cancers, similar findings have not been identified for microbiomes from other body sites and their correlation to treatment response and immune related adverse events (irAEs) in lung cancer (LC) patients receiving ICIs. METHODS We designed a prospective cohort study conducted from 2018 to 2020 at a single-center academic institution to assess for correlations between the microbiome in various body sites with treatment response and development of irAEs in LC patients treated with ICIs. Patients must have had measurable disease, ECOG 0-2, and good organ function to be included. Data was collected for analysis from January 2019 to October 2020. Patients with histopathologically confirmed, advanced/metastatic LC planned to undergo immunotherapy-based treatment were enrolled between September 2018 and June 2019. Nasal, buccal and gut microbiome samples were obtained prior to initiation of immunotherapy +/- chemotherapy, at development of adverse events (irAEs), and at improvement of irAEs to grade 1 or less. RESULTS Thirty-seven patients were enrolled, and 34 patients were evaluable for this report. 32 healthy controls (HC) from the same geographic region were included to compare baseline gut microbiota. Compared to HC, LC gut microbiota exhibited significantly lower α-diversity. The gut microbiome of patients who did not suffer irAEs were found to have relative enrichment of Bifidobacterium (p = 0.001) and Desulfovibrio (p = 0.0002). Responders to combined chemoimmunotherapy exhibited increased Clostridiales (p = 0.018) but reduced Rikenellaceae (p = 0.016). In responders to chemoimmunotherapy we also observed enrichment of Finegoldia in nasal microbiome, and increased Megasphaera but reduced Actinobacillus in buccal samples. Longitudinal samples exhibited a trend of α-diversity and certain microbial changes during the development and resolution of irAEs. CONCLUSIONS This pilot study identifies significant differences in the gut microbiome between HC and LC patients, and their correlation to treatment response and irAEs in LC. In addition, it suggests potential predictive utility in nasal and buccal microbiomes, warranting further validation with a larger cohort and mechanistic dissection using preclinical models. TRIAL REGISTRATION ClinicalTrials.gov, NCT03688347 . Retrospectively registered 09/28/2018.
Collapse
Affiliation(s)
- Justin Chau
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, USA
| | - Meeta Yadav
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, USA
| | - Ben Liu
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, USA
| | - Muhammad Furqan
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, USA
| | - Qun Dai
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, USA
| | - Shailesh Shahi
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, USA
| | - Arnav Gupta
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, USA
- Birla Institute of Technology and Science Pilani, KK Birla Goa Campus, Zuarinagar, India
| | - Keri Nace Mercer
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, USA
| | - Evan Eastman
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, USA
| | - Taher Abu Hejleh
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, USA
| | - Carlos Chan
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, USA
| | - George J Weiner
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, USA
| | | | - Sonny T M Lee
- Division of Biology, Kansas State University, Manhattan, USA
| | - Cuncong Zhong
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, USA
| | - Ashutosh Mangalam
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, USA
| | - Jun Zhang
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, USA.
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, USA.
- Department of Cancer Biology, University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, USA.
| |
Collapse
|
13
|
Wang N, Sun T, Xu J. Tumor-related Microbiome in the Breast Microenvironment and Breast Cancer. J Cancer 2021; 12:4841-4848. [PMID: 34234854 PMCID: PMC8247384 DOI: 10.7150/jca.58986] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 05/30/2021] [Indexed: 12/13/2022] Open
Abstract
Despite the significant progress in diagnosis and treatment over the past years in the understanding of breast cancer pathophysiology, it remains one of the leading causes of mortality worldwide among females. Novel technologies are needed to improve better diagnostic and therapeutic approaches, and to better understand the role of tumor-environment microbiome players involved in the progression of this disease. The gut environment is enriched with over 100 trillion microorganisms, which participate in metabolic diseases, obesity, and inflammation, and influence the response to therapy. In addition to the direct metabolic effects of the gut microbiome, accumulating evidence has revealed that a microbiome also exists in the breast and in breast cancer tissue. This microbiome enriched in the breast environment and the tumor microenvironment may modulate effects potentially associated with carcinogenesis and therapeutic interventions in breast tissue, which to date have not been properly acknowledged. Herein, we review the most recent works associated with the population dynamics of breast microbes and explore the significance of the microbiome on diagnosis, tumor development, response to chemotherapy, endocrine therapy, and immunotherapy. To overcome the low reproducibility of evaluations of tumor-related microbiome, sequencing technical escalation and machine deep learning algorithms may be valid for standardization of assessment for breast-related microbiome and their applications as powerful biomarkers for prognosis and predictive response in the future.
Collapse
Affiliation(s)
- Na Wang
- Department of Breast Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China, 110042
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China, 110042
| | - Tao Sun
- Department of Breast Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China, 110042
- Key Laboratory of Liaoning Breast Cancer Research, Shenyang, Liaoning, China
| | - Junnan Xu
- Department of Breast Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China, 110042
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China, 110042
| |
Collapse
|
14
|
Huang C, Li M, Liu B, Zhu H, Dai Q, Fan X, Mehta K, Huang C, Neupane P, Wang F, Sun W, Umar S, Zhong C, Zhang J. Relating Gut Microbiome and Its Modulating Factors to Immunotherapy in Solid Tumors: A Systematic Review. Front Oncol 2021; 11:642110. [PMID: 33816289 PMCID: PMC8012896 DOI: 10.3389/fonc.2021.642110] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/24/2021] [Indexed: 12/18/2022] Open
Abstract
Background: Gut microbiome is proved to affect the activity of immunotherapy in certain tumors. However, little is known if there is universal impact on both the treatment response and adverse effects (AEs) of immune checkpoint inhibitors (ICIs) across multiple solid tumors, and whether such impact can be modulated by common gut microbiome modifiers, such as antibiotics and diet. Methods: A systematic search in PubMed followed by stringent manual review were performed to identify clinical cohort studies that evaluated the relevance of gut microbiome to ICIs (response and/or AEs, 12 studies), or association of antibiotics with ICIs (17 studies), or impact of diet on gut microbiome (16 studies). Only original studies published in English before April 1st, 2020 were used. Qualified studies identified in the reference were also included. Results: At the phylum level, patients who had enriched abundance in Firmicutes and Verrucomicrobia almost universally had better response from ICIs, whereas those who were enriched in Proteobacteria universally presented with unfavorable outcome. Mixed correlations were observed for Bacteroidetes in relating to treatment response. Regarding the AEs, Firmicutes correlated to higher incidence whereas Bacteroidetes were clearly associated with less occurrence. Interestingly, across various solid tumors, majority of the studies suggested a negative association of antibiotic use with clinical response from ICIs, especially within 1-2 month prior to the initiation of ICIs. Finally, we observed a significant correlation of plant-based diet in relating to the enrichment of “ICI-favoring” gut microbiome (P = 0.0476). Conclusions: Gut microbiome may serve as a novel modifiable biomarker for both the treatment response and AEs of ICIs across various solid tumors. Further study is needed to understand the underlying mechanism, minimize the negative impact of antibiotics on ICIs, and gain insight regarding the role of diet so that this important lifestyle factor can be harnessed to improve the therapeutic outcomes of cancer immunotherapy partly through its impact on gut microbiome.
Collapse
Affiliation(s)
- Chengliang Huang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States
| | - Meizhang Li
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| | - Ben Liu
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, United States
| | - Huanbo Zhu
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States.,Department of Gastrointestinal Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qun Dai
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States
| | - Xianming Fan
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Kathan Mehta
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States
| | - Chao Huang
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States
| | - Prakash Neupane
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States
| | - Fen Wang
- Department of Radiation Oncology, University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, United States
| | - Weijing Sun
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States
| | - Shahid Umar
- Department of Surgery, University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, United States.,Department of Cancer Biology, University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, United States
| | - Cuncong Zhong
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, United States
| | - Jun Zhang
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States.,Department of Cancer Biology, University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
15
|
Martins Lopes MS, Machado LM, Ismael Amaral Silva PA, Tome Uchiyama AA, Yen CT, Ricardo ED, Mutao TS, Pimenta JR, Shimba DS, Hanriot RM, Peixoto RD. Antibiotics, cancer risk and oncologic treatment efficacy: a practical review of the literature. Ecancermedicalscience 2020; 14:1106. [PMID: 33144874 PMCID: PMC7581329 DOI: 10.3332/ecancer.2020.1106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Indexed: 12/12/2022] Open
Abstract
Antibiotics have been extensively used to treat infectious diseases over the past century and have largely contributed to increased life expectancy over time. However, antibiotic use can impose profound and protracted changes to the diversity of the microbial ecosystem, affecting the composition of up to 30% of the bacterial species in the gut microbiome. By modifying human microbiota composition, antibiotics alter the action of several oncologic drugs, potentially leading to decreased efficacy and increased toxicities. Whether antibiotics interfere with cancer therapies or even increase the risk of cancer development has been under investigation, and no randomised trials have been conducted so far. The aim of the current review is to describe the possible effects of antibiotic therapies on different oncologic treatments, especially immunotherapies, and to explore the link between previous antibiotics use and the development of cancer.
Collapse
Affiliation(s)
| | | | | | | | - Cheng T Yen
- Hospital Alemão Oswaldo Cruz, São Paulo, Brazil
| | | | | | | | | | | | - Renata D Peixoto
- Centro Paulista de Oncologia (Grupo Oncoclínicas), São Paulo, Brazil
| |
Collapse
|
16
|
Zhou H, Suo J, Zhu J. [Therapeutic Relevance of Human Microbiota and Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2019; 22:464-469. [PMID: 31315786 PMCID: PMC6712272 DOI: 10.3779/j.issn.1009-3419.2019.07.09] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
人体菌群与人类健康状态密切相关,如人体菌群的失调可能导致糖尿病、胃肠道疾病、肥胖等疾病的发生。人体内微生物与约20%的恶性肿瘤有关,肺癌(lung cancer, LC)是目前最为常见的恶性肿瘤之一,我国男性LC发病率及死亡率高居所有恶性肿瘤之首。近来研究表明,人体菌群可能通过代谢、炎症或免疫等途径影响着LC的发生,同时影响LC对放化疗、基因治疗、免疫治疗等治疗方法的疗效,如免疫治疗,是用于治疗LC的一种极有前景的手段,但不同患者从中获益不一,包含以肺癌细胞株的实验表明肠道微生物群可通过与宿主免疫系统的相互作用调节对免疫治疗的反应。但针对肺癌患者,肠道菌群是否仍能对免疫治疗进行调节仍存在争议。本文就人体菌群与LC的治疗相关性的近来研究进展进行综述。
Collapse
Affiliation(s)
- Huijie Zhou
- Department of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiaojiao Suo
- Department of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiang Zhu
- Department of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|