1
|
Bonnefin C, Schneider S, Gérard E, Dutriaux C, Ferte T, Prey S, Guicheney M, Ducharme O, Pedeboscq S, Beylot-Barry M, Pham-Ledard A. Antibiotics use decreases survival in cutaneous squamous cell carcinoma patients receiving immune checkpoint inhibitors. Eur J Cancer 2025; 217:115223. [PMID: 39874910 DOI: 10.1016/j.ejca.2025.115223] [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: 11/15/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICI) have become the first-line therapy in patients with advanced cutaneous squamous cell carcinoma (cSCC). Antibiotics (ATB) have been reported to reduce ICI response in cancers, but this has not been evaluated in cSCC. AIM To evaluate ATB exposure at the onset of ICI in cSCC patients and to analyze its impact on outcome. METHODS This single-center retrospective study included all patients who started anti-PD-1 for cSCC between March 2019 and July 2023. Exposure to ATB within 3 months prior and after the onset of ICI (ATB 3-3), including patients exposed within 1 month prior and after (ATB 1-1) were recorded. Response to ICI and survival were compared between patients with or without ATB exposure. RESULTS Among 104 patients included, 45 % were classified into ATB 3-3 subgroup, and 20 % to ATB 1-1. Disease control rate at 3 months were lower in both ATB 1-1 and ATB 3-3 subgroups, compared to their control group (p = 0.02 and 0.03, respectively). The overall survival and disease specific survival were lower in the ATB 1-1 subgroup, compared to control group (p = 0.04 and p = 0.01, respectively). Median progression free survival was 127 days in ATB 1-1 group, significantly lower than the control group (not reached), p = 0.005. CONCLUSION ATB intake was very frequent at ICI initiation in cSCC patients. In our cohort, ATB use within 1 month before or after ICI initiation significantly impacted survival, highlighting the need for caution when prescribing antibiotics in this population.
Collapse
Affiliation(s)
- C Bonnefin
- Department of Dermatology, CHU Bordeaux, Bordeaux F-33000, France
| | - S Schneider
- Department of Dermatology, CHU Bordeaux, Bordeaux F-33000, France
| | - E Gérard
- Department of Dermatology, CHU Bordeaux, Bordeaux F-33000, France
| | - C Dutriaux
- Department of Dermatology, CHU Bordeaux, Bordeaux F-33000, France; Bordeaux Institute of Oncology, INSERM U1312, Team Translational Research on Oncodermatology and Orphean Skin Diseases, Univ. Bordeaux, Bordeaux F-33000, France
| | - T Ferte
- Public Health Centre, Methodological Support Unit for Clinical and Epidemiological Research (USMR), CHU Bordeaux, Bordeaux F-33000, France
| | - S Prey
- Department of Dermatology, CHU Bordeaux, Bordeaux F-33000, France; Bordeaux Institute of Oncology, INSERM U1312, Team Translational Research on Oncodermatology and Orphean Skin Diseases, Univ. Bordeaux, Bordeaux F-33000, France
| | - M Guicheney
- Department of Dermatology, CHU Bordeaux, Bordeaux F-33000, France
| | - O Ducharme
- Department of Dermatology, CHU Bordeaux, Bordeaux F-33000, France
| | - S Pedeboscq
- Department of Pharmacy, CHU Bordeaux, Bordeaux F-33000, France
| | - M Beylot-Barry
- Department of Dermatology, CHU Bordeaux, Bordeaux F-33000, France; Bordeaux Institute of Oncology, INSERM U1312, Team Translational Research on Oncodermatology and Orphean Skin Diseases, Univ. Bordeaux, Bordeaux F-33000, France
| | - A Pham-Ledard
- Department of Dermatology, CHU Bordeaux, Bordeaux F-33000, France; Bordeaux Institute of Oncology, INSERM U1312, Team Translational Research on Oncodermatology and Orphean Skin Diseases, Univ. Bordeaux, Bordeaux F-33000, France.
| |
Collapse
|
2
|
Jiang SS, Kang ZR, Chen YX, Fang JY. The gut microbiome modulate response to immunotherapy in cancer. SCIENCE CHINA. LIFE SCIENCES 2025; 68:381-396. [PMID: 39235561 DOI: 10.1007/s11427-023-2634-7] [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: 04/14/2024] [Accepted: 06/05/2024] [Indexed: 09/06/2024]
Abstract
Gut microbiota have been reported to play an important role in the occurrence and development of malignant tumors. Currently, clinical studies have identified specific gut microbiota and its metabolites associated with efficacy of immunotherapy in multiple types of cancers. Preclinical investigations have elucidated that gut microbiota modulate the antitumor immunity and affect the efficacy of cancer immunotherapy. Certain microbiota and its metabolites may favorably remodel the tumor microenvironment by engaging innate and/or adaptive immune cells. Understanding how the gut microbiome interacts with cancer immunotherapy opens new avenues for improving treatment strategies. Fecal microbial transplants, probiotics, dietary interventions, and other strategies targeting the microbiota have shown promise in preclinical studies to enhance the immunotherapy. Ongoing clinical trials are evaluating these approaches. This review presents the recent advancements in understanding the dynamic interplay among the host immunity, the microbiome, and cancer immunotherapy, as well as strategies for modulating the microbiome, with a view to translating into clinical applications.
Collapse
Affiliation(s)
- Shan-Shan Jiang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China
| | - Zi-Ran Kang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China
| | - Ying-Xuan Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China.
| |
Collapse
|
3
|
Radoš L, Golčić M, Mikolašević I. The Relationship Between the Modulation of Intestinal Microbiota and the Response to Immunotherapy in Patients with Cancer. Biomedicines 2025; 13:96. [PMID: 39857680 PMCID: PMC11761299 DOI: 10.3390/biomedicines13010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
The intestinal microbiota is an important part of the human body, and its composition can affect the effectiveness of immunotherapy. In the last few years, the modulation of intestinal microbiota in order to improve the effectiveness of immunotherapy has become a current topic in the scientific community, but there is a lack of research in this area. In this review, the goal was to analyze the current relevant literature related to the modulation of intestinal microbiota and the effectiveness of immunotherapy in the treatment of cancer. The effects of antibiotics, probiotics, diet, and fecal microbial transplantation were analyzed separately. It was concluded that the use of antibiotics, especially broad-spectrum types or larger quantities, causes dysbiosis of the intestinal microbiota, which can reduce the effectiveness of immunotherapy. While dysbiosis could be repaired by probiotics and thus improve the effectiveness of immunotherapy, the use of commercial probiotics without evidence of intestinal dysbiosis has not yet been sufficiently tested to confirm its safety for cancer for immunotherapy-treated cancer patients. A diet consisting of sufficient amounts of fiber, as well as a diet with higher salt content positively correlates with the success of immunotherapy. Fecal transplantation is a safe and realistic adjuvant option for the treatment of cancer patients with immunotherapy, but more clinical trials are necessary. Modulating the microbiota composition indeed changes the effectiveness of immunotherapy, but in the future, more human studies should be organized to precisely determine the types and procedures of microbiota modulation.
Collapse
Affiliation(s)
- Laura Radoš
- Department for Emergency Medicine of Primorsko-Goranska County, 51000 Rijeka, Croatia;
| | - Marin Golčić
- Clinic for Tumors, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia;
- School of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Ivana Mikolašević
- Clinic for Tumors, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia;
- School of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
4
|
Li C, Chen Z, Shi J, Zheng X. Efficacy of Ambroxol Combined with Loquat Syrup on Bacterial Pneumonia in Mice. J Inflamm Res 2024; 17:10107-10117. [PMID: 39639928 PMCID: PMC11619114 DOI: 10.2147/jir.s478655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
Purpose Bacterial pneumonia is a prevalent respiratory disease and a primary cause of death among hospitalized patients. Ambroxol and loquat syrup are widely utilized pharmaceuticals for managing respiratory infections in China. This study investigates the potential application and efficacy of combining ambroxol with loquat syrup for treating bacterial pneumonia. Methods In this study, mice with P. aeruginosa-induced bacterial pneumonia were used to evaluate the therapeutic effects of ambroxol, loquat syrup, and their combination. A bacterial plate coating assay was performed to measure the P. aeruginosa content in saliva, lung tissue, and bronchoalveolar lavage fluid (BALF). A plate colony counting assay was conducted to assess the antibacterial activity of ambroxol and loquat syrup. Serum, BALF, and lung tissues were analyzed using qPCR, ELISA, immunohistochemistry, and hematoxylin-eosin staining to evaluate disease severity. Results In this study, the experimental results demonstrate that, compared to treatment with ambroxol and/or loquat syrup alone, the combined administration of ambroxol and loquat syrup significantly increases the volume of saliva expectorated by mice infected with bacteria, concurrently augmenting bacterial presence in saliva while diminishing bacterial burden in the lungs, with significant differences observed (p<0.05). Furthermore, the combined therapy of ambroxol and loquat syrup achieved better therapeutic effects on P. aeruginosa pneumonia compared to ambroxol and/or loquat syrup alone (p<0.05), as evidenced by significantly reduced P. aeruginosa-induced lung injury, improved lung permeability, decreased inflammatory cell infiltration, and lower expression of inflammatory cytokines. Conclusion These findings suggest that the combination therapy of ambroxol and loquat syrup presents a safe and feasible new treatment strategy for bacterial pneumonia, offering promising benefits for ameliorating lung tissue damage and inflammation.
Collapse
Affiliation(s)
- Chenping Li
- Department of Pulmonary and Critical Care Medicine (PCCM), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
| | - Zhi Chen
- Department of Pulmonary and Critical Care Medicine (PCCM), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
| | - Jiaqi Shi
- Department of Pulmonary and Critical Care Medicine (PCCM), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
| | - Xiuyun Zheng
- Department of Pulmonary and Critical Care Medicine (PCCM), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
| |
Collapse
|
5
|
Hrubesz G, Leigh J, Ng TL. Understanding the relationship between breast cancer, immune checkpoint inhibitors, and gut microbiota: a narrative review. TRANSLATIONAL BREAST CANCER RESEARCH : A JOURNAL FOCUSING ON TRANSLATIONAL RESEARCH IN BREAST CANCER 2024; 5:31. [PMID: 39534584 PMCID: PMC11557166 DOI: 10.21037/tbcr-24-14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 08/27/2024] [Indexed: 11/16/2024]
Abstract
Background and Objective The composition of gut microbiota plays an important role in predicting and influencing outcomes of cancer treated with immunotherapy. Our objective is to summarize the role of gut microbiota and immunotherapy in breast cancer. Methods A systematic search from inception until July 2024 of key search terms including immunity, breast neoplasm, gastrointestinal microbiome/microbiota, fecal microbiota transplantation, pro- and prebiotics, antibiotics and immunotherapy using EMBASE, MEDLINE and CENTRAL was conducted. The results were screened by two reviewers independently and synthesized and presented descriptively. Key Content and Findings Thirteen studies (5 clinical, 8 pre-clinical) met the eligibility criteria and were published from 2020-2024. Clinical studies showed that the composition and diversity of gut microbiota was associated with patient response to immunotherapy. In pre-clinical studies, dysbiotic states induced by obesity, antibiotics, and diet were associated with immunosuppression and influenced response to programmed cell death-ligand 1 (PD-L1) inhibitors. Microbiota-modulating treatments such as probiotics showed the ability to enhance response to immunotherapy, indicating their potential use as adjunct therapies in breast cancer treatment. Conclusions The composition of gut microbiota could help predict the chance of response to immunotherapy, and modulating gut microbiota has the potential to enhance the efficacy of chemo-immunotherapy in breast cancer. However, the available data relating to breast cancer are limited. Larger prospective studies are required to further elucidate their role as a biomarker and treatment.
Collapse
Affiliation(s)
- Gabriella Hrubesz
- Division of Medical Oncology, The Ottawa Hospital Cancer Center, Ottawa, Ontario, Canada
| | - Jennifer Leigh
- Division of Medical Oncology, The Ottawa Hospital Cancer Center, Ottawa, Ontario, Canada
| | - Terry L Ng
- Division of Medical Oncology, The Ottawa Hospital Cancer Center, Ottawa, Ontario, Canada
| |
Collapse
|
6
|
Liu C, Fu L, Wang Y, Yang W. Influence of the gut microbiota on immune cell interactions and cancer treatment. J Transl Med 2024; 22:939. [PMID: 39407240 PMCID: PMC11476117 DOI: 10.1186/s12967-024-05709-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
The tumour microenvironment represents a novel frontier in oncological research. Over the past decade, accumulating evidence has underscored the importance of the tumour microenvironment (TME), including tumour cells, stromal cells, immune cells, and various secreted factors, which collectively influence tumour growth, invasion, and responses to therapeutic agents. Immune cells within the TME are now widely acknowledged to play pivotal roles in tumour development and treatment. While some perspectives have posited that immune cells within the TME facilitate tumour progression and confer resistance to therapeutic interventions, contrasting conclusions also exist. Affirmative and negative conclusions appear to be context dependent, and a unified consensus has yet to be reached. The burgeoning body of research on the relationship between the gut microbiota and tumours in recent years has led to a growing understanding. Most studies have indicated that specific components of the gut microbiota, such as unique bacterial communities or specific secretory factors, play diverse roles in regulating immune cells within the TME, thereby influencing the prognosis and outcomes of cancer treatments. A detailed understanding of these factors could provide novel insights into the TME and cancer therapy. In this study, we aimed to synthesise information on the interactions between the gut microbiota and immune cells within the TME, providing an in-depth exploration of the potential guiding implications for future cancer therapies.
Collapse
Affiliation(s)
- Chunxiao Liu
- Department of Gastroenterological Surgery, Hengqin Hospital, First Affiliated Hospital of Guangzhou Medical University, No. 118 Baoxing Road, Hengqin, Guangdong, 519031, China
| | - Lingfeng Fu
- Department of Gastroenterological Surgery, Hengqin Hospital, First Affiliated Hospital of Guangzhou Medical University, No. 118 Baoxing Road, Hengqin, Guangdong, 519031, China
| | - Yuxin Wang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, No. 1838, North Guangzhou Avenue, Guangzhou, Guangdong, 510515, China.
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Weijun Yang
- Department of Gastroenterological Surgery, Hengqin Hospital, First Affiliated Hospital of Guangzhou Medical University, No. 118 Baoxing Road, Hengqin, Guangdong, 519031, China.
| |
Collapse
|
7
|
Haddad A, Holder AM. Microbiome and Immunotherapy for Melanoma: Are We Ready for Clinical Application? Hematol Oncol Clin North Am 2024; 38:1061-1070. [PMID: 38908958 DOI: 10.1016/j.hoc.2024.05.010] [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] [Indexed: 06/24/2024]
Abstract
The microbiome plays a substantial role in the efficacy of immune checkpoint blockade (ICB) in patients with metastatic melanoma. While the exact gut microbiome composition and the pathways involved in this interaction are not clearly delineated, novel studies and ongoing clinical trials are likely to reveal findings applicable to the clinical setting for the prediction and optimization of response to ICB. Nevertheless, lifestyle modifications, including high fiber diet, avoidance of unnecessary antibiotic prescriptions, and careful use of probiotics may be helpful to optimize the "health" of the gut microbiome and potentially enhance response to ICB in patients with melanoma.
Collapse
Affiliation(s)
- Antony Haddad
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1484, Houston, TX 77030, USA. https://twitter.com/Haddad_Antony
| | - Ashley M Holder
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1484, Houston, TX 77030, USA.
| |
Collapse
|
8
|
Ji G, Zhao J, Si X, Song W. Targeting bacterial metabolites in tumor for cancer therapy: An alternative approach for targeting tumor-associated bacteria. Adv Drug Deliv Rev 2024; 211:115345. [PMID: 38834140 DOI: 10.1016/j.addr.2024.115345] [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: 02/03/2024] [Revised: 05/11/2024] [Accepted: 05/29/2024] [Indexed: 06/06/2024]
Abstract
Emerging evidence reveal that tumor-associated bacteria (TAB) can facilitate the initiation and progression of multiple types of cancer. Recent work has emphasized the significant role of intestinal microbiota, particularly bacteria, plays in affecting responses to chemo- and immuno-therapies. Hence, it seems feasible to improve cancer treatment outcomes by targeting intestinal bacteria. While considering variable richness of the intestinal microbiota and diverse components among individuals, direct manipulating the gut microbiota is complicated in clinic. Tumor initiation and progression requires the gut microbiota-derived metabolites to contact and reprogram neoplastic cells. Hence, directly targeting tumor-associated bacteria metabolites may have the potential to provide alternative and innovative strategies to bypass the gut microbiota for cancer therapy. As such, there are great opportunities to explore holistic approaches that incorporates TAB-derived metabolites and related metabolic signals modulation for cancer therapy. In this review, we will focus on key opportunistic areas by targeting TAB-derived metabolites and related metabolic signals, but not bacteria itself, for cancer treatment, and elucidate future challenges that need to be addressed in this emerging field.
Collapse
Affiliation(s)
- Guofeng Ji
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Jingjing Zhao
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453100, China
| | - Xinghui Si
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China.
| |
Collapse
|
9
|
Alotaibi FM, Albalawi IAS, Anis AM, Alotaibi H, Khashwayn S, Alshammari K, Al-Tawfiq JA. The impact of antibiotic use in gastrointestinal tumors treated with immune checkpoint inhibitors: systematic review and meta-analysis. Front Med (Lausanne) 2024; 11:1415093. [PMID: 38887674 PMCID: PMC11180829 DOI: 10.3389/fmed.2024.1415093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
Background Immune checkpoint inhibitors (ICI) have improved overall survival in patients with different cancer types. However, treatment efficacy varies between patients depending on several factors. Recent research suggested that antibiotic-induced dysbiosis can impair ICI efficacy. Here we review the impact of antibiotic use in clinical outcome of patients with gastrointestinal cancer treated with ICI. Methods This is a systematic review and utilized a thorough search of MEDLINE, Cochrane, Scopus, EB-SCO, Web of Science of studies published till September 2023. The aim of the study is to determine the association between antibiotic use and ICI treatment efficacy in patients with gastrointestinal cancers (GI). We utilized a meta-analysis of the association between the use of antibiotics and overall survival and progression-free survival. Results Nine studies met the inclusion criteria with a total of 2,214 patients. The most common type of cancers was hepatocellular carcinoma (HCC). The majority of the studies were retrospective, and one was collective of clinical trials. The use of antibiotics was associated with decreased both overall survival [haz-ard ratio (HR) 1.92, 95% confidence interval (CI) 1.41, 2.63] and progression-free survival [HR 1.81, 95% CI 1.29, 2.54]. Conclusion The use of antibiotics may affect clinical outcomes in patients with GI cancers treated with ICI. Further prospective studies are needed to improve the understanding of this phenomenon. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42023462172.
Collapse
Affiliation(s)
- Faizah M. Alotaibi
- College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, Alahsa, Saudi Arabia
- King Abdullah International Medical Research Center, Alahsa, Saudi Arabia
| | | | - Amna M. Anis
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Department of Biomedical Engineering, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Hawazin Alotaibi
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Seham Khashwayn
- King Saud Bin Abdulaziz University for Health Sciences, Alhasa, Saudi Arabia
| | - Kanan Alshammari
- Department of Oncology, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
| | - Jaffar A. Al-Tawfiq
- Department of Specialty Internal Medicine and Quality, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia
- Infectious Disease Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Infectious Disease Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
10
|
Townsend MJ, Benque IJ, Li M, Grover S. Review article: Contemporary management of gastrointestinal, pancreatic and hepatic toxicities of immune checkpoint inhibitors. Aliment Pharmacol Ther 2024; 59:1350-1365. [PMID: 38590108 DOI: 10.1111/apt.17980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/13/2023] [Accepted: 03/21/2024] [Indexed: 04/10/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are effective oncologic agents which frequently cause immune-related adverse events (irAEs) which can impact multiple organ systems. Onco-Gastroenterology is a novel and emerging subspecialty within gastroenterology focused on cancer treatment-related complications. Gastroenterologists must be prepared to identify and manage diverse immune-mediated toxicities including enterocolitis, hepatitis, pancreatitis and other ICI-induced toxicities. AIM To provide a narrative review of the epidemiology, diagnostic evaluation and management of checkpoint inhibitor-induced gastrointestinal and hepatic toxicities. METHODS We searched Cochrane and PubMed databases for articles published through August 2023. RESULTS Gastrointestinal and hepatic irAEs include most commonly enterocolitis and hepatitis, but also pancreatitis, oesophagitis, gastritis, motility disorders (gastroparesis) and other rarer toxicities. Guidelines from the National Comprehensive Cancer Network, American Society of Clinical Oncology and European Society for Medical Oncology, in combination with emerging cohort and clinical trial data, offer strategies for management of ICI toxicities. Evaluation of irAEs severity by formal classification and clinical stability, and a thorough workup for alternative etiologies which may clinically mimic irAEs underlie initial management. Treatments include corticosteroids, biologics and other immunosuppressive agents plus supportive care; decisions on dosing, timing and choice of steroid adjuncts and potential for subsequent checkpoint inhibitor dosing are nuanced and toxicity-specific. CONCLUSIONS Expanding clinical trial and cohort data have clarified the epidemiology and clinical characteristics of gastrointestinal, pancreatic and hepatic toxicities of ICIs. Guidelines, though valuable, remain based principally on retrospective cohort data. Quality prospective, controlled studies may refine algorithms for treatment and potential immunotherapy rechallenge.
Collapse
Affiliation(s)
- Matthew J Townsend
- Department of Medicine, Duke University Hospital, Durham, North Carolina, USA
| | - Isaac J Benque
- University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Michael Li
- University of California San Francisco School of Medicine, San Francisco, California, USA
- Division of Gastroenterology, University of California San Francisco Medical Center, San Francisco, California, USA
| | - Shilpa Grover
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
11
|
Adamiak K, Gaida VA, Schäfer J, Bosse L, Diemer C, Reiter RJ, Slominski AT, Steinbrink K, Sionkowska A, Kleszczyński K. Melatonin/Sericin Wound Healing Patches: Implications for Melanoma Therapy. Int J Mol Sci 2024; 25:4858. [PMID: 38732075 PMCID: PMC11084828 DOI: 10.3390/ijms25094858] [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: 03/28/2024] [Revised: 04/19/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Melatonin and sericin exhibit antioxidant properties and may be useful in topical wound healing patches by maintaining redox balance, cell integrity, and regulating the inflammatory response. In human skin, melatonin suppresses damage caused by ultraviolet radiation (UVR) which involves numerous mechanisms associated with reactive oxygen species/reactive nitrogen species (ROS/RNS) generation and enhancing apoptosis. Sericin is a protein mainly composed of glycine, serine, aspartic acid, and threonine amino acids removed from the silkworm cocoon (particularly Bombyx mori and other species). It is of interest because of its biodegradability, anti-oxidative, and anti-bacterial properties. Sericin inhibits tyrosinase activity and promotes cell proliferation that can be supportive and useful in melanoma treatment. In recent years, wound healing patches containing sericin and melatonin individually have attracted significant attention by the scientific community. In this review, we summarize the state of innovation of such patches during 2021-2023. To date, melatonin/sericin-polymer patches for application in post-operational wound healing treatment has been only sparingly investigated and it is an imperative to consider these materials as a promising approach targeting for skin tissue engineering or regenerative dermatology.
Collapse
Affiliation(s)
- Katarzyna Adamiak
- Department of Biomaterials and Cosmetic Chemistry, Faculty of Chemistry, Nicolaus Copernicus University, Gagarin 7, 87-100 Toruń, Poland; (K.A.); (A.S.)
| | - Vivian A. Gaida
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany; (V.A.G.); (J.S.); (L.B.); (C.D.); (K.S.)
| | - Jasmin Schäfer
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany; (V.A.G.); (J.S.); (L.B.); (C.D.); (K.S.)
| | - Lina Bosse
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany; (V.A.G.); (J.S.); (L.B.); (C.D.); (K.S.)
| | - Clara Diemer
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany; (V.A.G.); (J.S.); (L.B.); (C.D.); (K.S.)
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, Long School of Medicine, UT Health, San Antonio, TX 78229, USA;
| | - Andrzej T. Slominski
- Department of Dermatology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Pathology and Laboratory Medicine Service, VA Medical Center, Birmingham, AL 35294, USA
| | - Kerstin Steinbrink
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany; (V.A.G.); (J.S.); (L.B.); (C.D.); (K.S.)
| | - Alina Sionkowska
- Department of Biomaterials and Cosmetic Chemistry, Faculty of Chemistry, Nicolaus Copernicus University, Gagarin 7, 87-100 Toruń, Poland; (K.A.); (A.S.)
| | - Konrad Kleszczyński
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany; (V.A.G.); (J.S.); (L.B.); (C.D.); (K.S.)
| |
Collapse
|
12
|
Kulkarni AA, Jain A, Jewett PI, Desai N, Van 't Veer L, Hirst G, Yee D, Blaes AH. Association of antibiotic exposure with residual cancer burden in HER2-negative early stage breast cancer. NPJ Breast Cancer 2024; 10:24. [PMID: 38531875 PMCID: PMC10966095 DOI: 10.1038/s41523-024-00630-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Antibiotic exposure during immunotherapy (IO) has been shown to negatively affect clinical outcomes in various cancer types. The aim of this study was to evaluate whether antibiotic exposure in patients with high-risk early-stage HER2-negative breast cancer (BC) undergoing treatment with neoadjuvant pembrolizumab impacted residual cancer burden (RCB) and pathologic complete response (pCR) in the pembrolizumab-4 arm of the ISPY-2 clinical trial. Patients received pembrolizumab for four cycles concurrently with weekly paclitaxel for 12 weeks, followed by four cycles of doxorubicin plus cyclophosphamide every 2 or 3 weeks. Patients who received at least one dose of systemic antibiotics concurrently at the time of immunotherapy (IO) were included in the antibiotic exposure group (ATB+). All other participants were included in the control group (ATB-). RCB index and PCR rates were compared between the ATB+ and ATB- groups using t-tests and Chi-squared tests, and linear and logistic regression models, respectively. Sixty-six patients were included in the analysis. 18/66 (27%) patients were in the ATB+ group. Antibiotic use during IO was associated with a higher mean RCB index (1.80 ± 1.43 versus 1.08 ± 1.41) and a lower pCR rate (27.8% versus 52.1%). The association between antibiotic use and the RCB index remained significant in multivariable linear regression analysis (RCB index-coefficient 0.86, 95% CI 0.20-1.53, P = 0.01). Our findings suggest that concurrent antibiotic exposure during neoadjuvant pembrolizumab in HER2-negative early-stage BC is associated with higher RCB. Further validation in larger cohorts is needed to confirm these findings.
Collapse
Affiliation(s)
- Amit A Kulkarni
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, USA.
| | - Aditya Jain
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Patricia I Jewett
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Nidhi Desai
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Laura Van 't Veer
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Gillian Hirst
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Douglas Yee
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Anne H Blaes
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
13
|
Liu B, Liu Z, Jiang T, Gu X, Yin X, Cai Z, Zou X, Dai L, Zhang B. Univariable and multivariable Mendelian randomization study identified the key role of gut microbiota in immunotherapeutic toxicity. Eur J Med Res 2024; 29:161. [PMID: 38475836 DOI: 10.1186/s40001-024-01741-7] [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: 09/25/2023] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND In cancer patients receiving immune checkpoint inhibitors (ICIs), there is emerging evidence suggesting a correlation between gut microbiota and immune-related adverse events (irAEs). However, the exact roles of gut microbiota and the causal associations are yet to be clarified. METHODS To investigate this, we first conducted a univariable bi-directional two-sample Mendelian randomization (MR) analysis. Instrumental variables (IVs) for gut microbiota were retrieved from the MiBioGen consortium (18,340 participants). GWAS summary data for irAEs were gathered from an ICIs-treated cohort with 1,751 cancer patients. Various MR analysis methods, including inverse variance weighted (IVW), MR PRESSO, maximum likelihood (ML), weighted median, weighted mode, and cML-MA-BIC, were used. Furthermore, multivariable MR (MVMR) analysis was performed to account for possible influencing instrumental variables. RESULTS Our analysis identified fourteen gut bacterial taxa that were causally associated with irAEs. Notably, Lachnospiraceae was strongly associated with an increased risk of both high-grade and all-grade irAEs, even after accounting for the effect of BMI in the MVMR analysis. Akkermansia, Verrucomicrobiaceae, and Anaerostipes were found to exert protective roles in high-grade irAEs. However, Ruminiclostridium6, Coprococcus3, Collinsella, and Eubacterium (fissicatena group) were associated with a higher risk of developing high-grade irAEs. RuminococcaceaeUCG004, and DefluviitaleaceaeUCG011 were protective against all-grade irAEs, whereas Porphyromonadaceae, Roseburia, Eubacterium (brachy group), and Peptococcus were associated with an increased risk of all-grade irAEs. CONCLUSIONS Our analysis highlights a strong causal association between Lachnospiraceae and irAEs, along with some other gut microbial taxa. These findings provide potential modifiable targets for managing irAEs and warrant further investigation.
Collapse
Affiliation(s)
- Baike Liu
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Zheran Liu
- Department of Biotherapy and National Clinical Research Center for Geriatrics, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Tianxiang Jiang
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xiangshuai Gu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xiaonan Yin
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Zhaolun Cai
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xiaoqiao Zou
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Bo Zhang
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
14
|
Elkrief A, Waters NR, Smith N, Dai A, Slingerland J, Aleynick N, Febles B, Gogia P, Socci ND, Lumish M, Giardina PA, Chaft JE, Eng J, Motzer RJ, Mendelsohn RB, Markey KA, Zhuang M, Li Y, Yang Z, Hollmann TJ, Rudin CM, van den Brink MR, Shia J, DeWolf S, Schoenfeld AJ, Hellmann MD, Babady NE, Faleck DM, Peled JU. Immune-Related Colitis Is Associated with Fecal Microbial Dysbiosis and Can Be Mitigated by Fecal Microbiota Transplantation. Cancer Immunol Res 2024; 12:308-321. [PMID: 38108398 PMCID: PMC10932930 DOI: 10.1158/2326-6066.cir-23-0498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/08/2023] [Accepted: 12/15/2023] [Indexed: 12/19/2023]
Abstract
Colitis induced by treatment with immune-checkpoint inhibitors (ICI), termed irColitis, is a substantial cause of morbidity complicating cancer treatment. We hypothesized that abnormal fecal microbiome features would be present at the time of irColitis onset and that restoring the microbiome with fecal transplant from a healthy donor would mitigate disease severity. Herein, we present fecal microbiota profiles from 18 patients with irColitis from a single center, 5 of whom were treated with healthy-donor fecal microbial transplantation (FMT). Although fecal samples collected at onset of irColitis had comparable α-diversity to that of comparator groups with gastrointestinal symptoms, irColitis was characterized by fecal microbial dysbiosis. Abundances of Proteobacteria were associated with irColitis in multivariable analyses. Five patients with irColitis refractory to steroids and biologic anti-inflammatory agents received healthy-donor FMT, with initial clinical improvement in irColitis symptoms observed in four of five patients. Two subsequently exhibited recurrence of irColitis symptoms following courses of antibiotics. Both received a second "salvage" FMT that was, again, followed by clinical improvement of irColitis. In summary, we observed distinct microbial community changes that were present at the time of irColitis onset. FMT was followed by clinical improvements in several cases of steroid- and biologic-agent-refractory irColitis. Strategies to restore or prevent microbiome dysbiosis in the context of immunotherapy toxicities should be further explored in prospective clinical trials.
Collapse
Affiliation(s)
- Arielle Elkrief
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nicholas R. Waters
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Natalie Smith
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Angel Dai
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - John Slingerland
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nathan Aleynick
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Binita Febles
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Pooja Gogia
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nicholas D. Socci
- Bioinformatics Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Melissa Lumish
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Paul A. Giardina
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jamie E. Chaft
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Juliana Eng
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Robert J. Motzer
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Robin B. Mendelsohn
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Kate A. Markey
- Fred Hutchinson Cancer Center, Seattle, Washington; Division of Medical Oncology, University of Washington, Seattle, Washington
| | - Mingqiang Zhuang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yanyun Li
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Zhifan Yang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Travis J. Hollmann
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Bristol Myers Squibb, Princeton, NJ, USA
| | - Charles M. Rudin
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Marcel R.M. van den Brink
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jinru Shia
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Susan DeWolf
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Adam J. Schoenfeld
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Matthew D. Hellmann
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - N. Esther Babady
- Clinical Microbiology Service, Department of Pathology and Laboratory Medicine and the Infectious Disease Service, Department of Medicine Memorial Sloan Kettering Cancer Center, New York, NY
| | - David M. Faleck
- Gastroenterology, Hepatology & Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Jonathan U. Peled
- Weill Cornell Medical College, New York, NY
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
15
|
Chorti E, Kowall B, Hassel JC, Schilling B, Sachse M, Gutzmer R, Loquai C, Kähler KC, Hüsing A, Gilde C, Thielmann CM, Zaremba-Montenari A, Placke JM, Gratsias E, Martaki A, Roesch A, Ugurel S, Schadendorf D, Livingstone E, Stang A, Zimmer L. Association of antibiotic treatment with survival outcomes in treatment-naïve melanoma patients receiving immune checkpoint blockade. Eur J Cancer 2024; 200:113536. [PMID: 38306840 DOI: 10.1016/j.ejca.2024.113536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/29/2023] [Accepted: 01/04/2024] [Indexed: 02/04/2024]
Abstract
PURPOSE The interaction of gut microbiome and immune system is being studied with increasing interest. Disturbing factors, such as antibiotics may impact the immune system via gut and interfere with tumor response to immune checkpoint blockade (ICB). METHODS In this multicenter retrospective cohort study exclusively treatment-naïve patients with cutaneous or mucosal melanoma treated with first-line anti-PD-1 based ICB for advanced, non-resectable disease between 06/2013 and 09/2018 were included. Progression-free (PFS), and overall survival (OS) according to antibiotic exposure (within 60 days prior to ICB and after the start of ICB vs. no antibiotic exposure) were analyzed. To account for immortal time bias, data from patients with antibiotics during ICB were analyzed separately in the time periods before and after start of antibiotics. RESULTS Among 578 patients with first-line anti-PD1 based ICB, 7% of patients received antibiotics within 60 days prior to ICB and 19% after starting ICB. Antibiotic exposure prior to ICB was associated with worse PFS (adjusted HR 1.75 [95% CI 1.22-2.52]) and OS (adjusted HR 1.64 [95% CI 1.04-2.58]) by multivariate analysis adjusting for potential confounders. The use of antibiotics after the start of ICB had no effect on either PFS (adjusted HR 1.19; 95% CI 0.89-1.60) or OS (adjusted HR 1.08; 95% CI 0.75-1.57). CONCLUSIONS Antibiotic exposure within 60 days prior to ICB seems to be associated with worse PFS and OS in melanoma patients receiving first-line anti-PD1 based therapy, whereas antibiotics after the start of ICB do not appear to affect PFS or OS.
Collapse
Affiliation(s)
- Eleftheria Chorti
- Department of Dermatology, Essen University Hospital, West German Cancer Center, University of Duisburg-Essen and the German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Bernd Kowall
- Institute of Medical Informatics, Biometry and Epidemiology (IMIBE), University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Jessica C Hassel
- Skin Cancer Center, Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| | - Bastian Schilling
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Michael Sachse
- Department of Dermatology, Allergology and Phlebology, Bremerhaven Reinkenheide Hospital, Bremerhaven, Germany
| | - Ralf Gutzmer
- Department of Dermatology, Skin Cancer Center Hannover, Hannover Medical School, Hannover and Johannes Wesling Medical Center Ruhr University Bochum, Minden, Germany
| | - Carmen Loquai
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Katharina C Kähler
- Department of Dermatology, University Hospital Schleswig-Holstein, Campus Kiel, Rosalind-Franklin-Str. 7, 24105 Kiel, Germany
| | - Anika Hüsing
- Institute of Medical Informatics, Biometry and Epidemiology (IMIBE), University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Catharina Gilde
- Department of Dermatology, Skin Cancer Center Hannover, Hannover Medical School, Hannover and Johannes Wesling Medical Center Ruhr University Bochum, Minden, Germany
| | - Carl M Thielmann
- Department of Dermatology, Essen University Hospital, West German Cancer Center, University of Duisburg-Essen and the German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Anne Zaremba-Montenari
- Department of Dermatology, Essen University Hospital, West German Cancer Center, University of Duisburg-Essen and the German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Jan-Malte Placke
- Department of Dermatology, Essen University Hospital, West German Cancer Center, University of Duisburg-Essen and the German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Emmanouil Gratsias
- Department of Dermatology, Essen University Hospital, West German Cancer Center, University of Duisburg-Essen and the German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Anna Martaki
- Department of Dermatology, Essen University Hospital, West German Cancer Center, University of Duisburg-Essen and the German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Alexander Roesch
- Department of Dermatology, Essen University Hospital, West German Cancer Center, University of Duisburg-Essen and the German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Selma Ugurel
- Department of Dermatology, Essen University Hospital, West German Cancer Center, University of Duisburg-Essen and the German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Dirk Schadendorf
- Department of Dermatology, Essen University Hospital, West German Cancer Center, University of Duisburg-Essen and the German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany; National Center for Tumor Diseases (NCT)-West, Campus Essen, & Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Essen, Germany
| | - Elisabeth Livingstone
- Department of Dermatology, Essen University Hospital, West German Cancer Center, University of Duisburg-Essen and the German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Andreas Stang
- Institute of Medical Informatics, Biometry and Epidemiology (IMIBE), University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Lisa Zimmer
- Department of Dermatology, Essen University Hospital, West German Cancer Center, University of Duisburg-Essen and the German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany.
| |
Collapse
|
16
|
Mok K, Wu C, Chan S, Wong G, Wong VWS, Ma B, Lui R. Clinical Management of Gastrointestinal and Liver Toxicities of Immune Checkpoint Inhibitors. Clin Colorectal Cancer 2024; 23:4-13. [PMID: 38172003 DOI: 10.1016/j.clcc.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024]
Abstract
Immune checkpoint inhibitors have transformed the treatment paradigm for various types of cancer. Nonetheless, with the utilization of these groundbreaking treatments, immune-related adverse events (irAEs) are increasingly encountered. Colonic and hepatic involvement are among the most frequently encountered irAEs. Drug-induced side effects, infectious causes, and tumor-related symptoms are the key differentials for irAE complications. Potential risk factors for the development of irAEs include combination use of immune checkpoint inhibitors, past development of irAEs with other immunotherapy treatments, certain concomitant drugs, and a pre-existing personal or family history of autoimmune illness such as inflammatory bowel disease. The importance of early recognition, timely and proper management cannot be understated, as there are profound clinical implications on the overall cancer treatment plan and prognosis once these adverse events occur. Herein, we cover the clinical management of the well-established gastrointestinal irAEs of enterocolitis and hepatitis, and also provide an overview of several other emerging entities.
Collapse
Affiliation(s)
- Kevin Mok
- Department of Clinical Oncology, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Claudia Wu
- Division of Gastroenterology and Hepatology, Department of Medicine and Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong SAR, China; Institute of Digestive Disease, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Stephen Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir YK Pao Centre for Cancer, Hong Kong Cancer Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Grace Wong
- Division of Gastroenterology and Hepatology, Department of Medicine and Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong SAR, China; Institute of Digestive Disease, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Vincent Wai-Sun Wong
- Division of Gastroenterology and Hepatology, Department of Medicine and Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong SAR, China; Institute of Digestive Disease, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Brigette Ma
- Department of Clinical Oncology, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong SAR, China; Institute of Digestive Disease, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Rashid Lui
- Department of Clinical Oncology, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong SAR, China; Division of Gastroenterology and Hepatology, Department of Medicine and Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong SAR, China; Institute of Digestive Disease, Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
17
|
Wang J, Ma Y, Lin H, Wang J, Cao B. Predictive biomarkers for immune-related adverse events in cancer patients treated with immune-checkpoint inhibitors. BMC Immunol 2024; 25:8. [PMID: 38267897 PMCID: PMC10809515 DOI: 10.1186/s12865-024-00599-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 01/08/2024] [Indexed: 01/26/2024] Open
Abstract
PURPOSE The objective of this study was to identify potential predictors of immune-related adverse events (irAEs) in cancer patients receiving immune checkpoint inhibitor therapy among serum indexes, case data, and liquid biopsy results. METHODS We retrospectively analyzed 418 patients treated with anti-programmed cell death 1(PD-1)/PD-1 ligand (PD-L1) inhibitors from January 2018 to May 2022 in our cancer center. We identified factors that correlated with the occurrence of irAEs and evaluated associations between irAEs and anti-PD-1/PD-L1 inhibitor responses. RESULTS The incidence of irAEs was 42.1%, and pneumonitis (9.1%), thyroid toxicity (9.1%), cardiotoxicity (8.1%), and dermatologic toxicity (6.9%) were the four most common irAEs. Multivariate logistic analysis identified female sex, antibiotic use, higher post-treatment neutrophil-to-lymphocyte ratio (NLR), and higher baseline circulating tumor cell (CTC) level, as predictive biomarkers for the occurrence of irAEs. A lower baseline prognostic nutritional index (PNI), body mass index (BMI) ≥ 25 kg/m2, and higher post-treatment lactate dehydrogenase (LDH) level were predictive factors for more severe irAEs (higher severity grade). Patients without irAEs had better overall survival than those with irAEs. Specifically, pneumonitis and cardiotoxicity were found to be significant predictors of poor prognosis in the irAE subgroup with different organ-related irAEs. Low-dose steroid (dexamethasone 10 mg) treatment had no significant effect on outcomes. CONCLUSIONS Gender, antibiotic use, post-treatment NLR, and baseline CTC level are potential predictive biomarkers of irAEs, while baseline PNI, BMI, and post-treatment LDH may predict the severity of irAEs. The predictive effect of irAE occurrence on survival benefit may depend on the type of irAE.
Collapse
Affiliation(s)
- Jingting Wang
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, #95 Yong An Road, Xicheng District, Beijing, China
| | - Yan Ma
- Radiotherapy Department, Shijingshan Teaching Hospital of Capital Medical University Beijing, #24 Shijingshan Road, Shijingshan District, Beijing, 100040, China
| | - Haishan Lin
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, #95 Yong An Road, Xicheng District, Beijing, China
| | - Jing Wang
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, #95 Yong An Road, Xicheng District, Beijing, China.
| | - Bangwei Cao
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, #95 Yong An Road, Xicheng District, Beijing, China.
| |
Collapse
|
18
|
Zhong J, Xiong D, Liu Y, Yuan S. Association of antibiotic exposure with survival in patients with extensive-stage small cell lung cancer receiving immune checkpoint inhibitor therapy. Thorac Cancer 2024; 15:152-162. [PMID: 38010059 PMCID: PMC10788467 DOI: 10.1111/1759-7714.15172] [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: 09/04/2023] [Revised: 11/09/2023] [Accepted: 11/11/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have dramatically shifted the therapeutic paradigm of extensive-stage small cell lung cancer (ES-SCLC). Antibiotic (ATB) exposure before or during ICI therapy can harm the integrity of the gut microbiome and lead to intestinal dysbiosis, which has a profoundly negative impact on the treatment response for various malignancies. Whether this is applicable to ES-SCLC remains unclear. METHODS We retrospectively reviewed the electronic medical records of all patients diagnosed with ES-SCLC who were treated with ICI-based immunotherapies from July 2019 to December 2020 at Shandong Cancer Hospital and Institute, China. Outcomes with the use of ATBs before or after the first infusion of ICI, including progression-free survival (PFS) and overall survival (OS), were investigated using the Kaplan-Meier method. Multivariate analyses were also conducted using a Cox proportional hazards model. RESULTS A total of 214 patients were included, among whom 41 (19.2%) received ATBs within 2 months before or after the first initiation of ICI therapy and were assigned to the ATB group. The ATB group showed a shorter median PFS (4.3 vs. 6.3 months; HR = 1.43, 95% CI: 0.97-2.11; p = 0.043) and a significantly shorter median OS (6.9 vs. 13 months; HR = 1.47, 95% CI: 0.98-2.20; p = 0.033) than the non-ATB group. In the multivariate analysis, ATB exposure was markedly associated with worse PFS (HR = 1.47, 95% CI: 1.03-2.09, p = 0.035) and OS (HR = 1.46, 95% CI: 1.01-2.11, p = 0.043). CONCLUSIONS Our results demonstrate that ATB exposure was significantly associated with worse survival in ES-SCLC patients who received ICI therapy.
Collapse
Affiliation(s)
- Jiaqi Zhong
- Clinical Medical CollegeSouthwest Medical UniversityLuzhouChina
- Department of Radiation OncologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Dali Xiong
- Clinical Medical CollegeSouthwest Medical UniversityLuzhouChina
- Department of Radiation OncologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Yu Liu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University Digestive Endoscopy CenterJinanChina
| | - Shuanghu Yuan
- Clinical Medical CollegeSouthwest Medical UniversityLuzhouChina
- Department of Radiation OncologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
- Department of Radiation OncologyThe Affiliated Cancer Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
19
|
Götz L, Ansafi T, Gerken M, Klinkhammer-Schalke M, Fischl A, Riemenschneider MJ, Proescholdt M, Bumes E, Kölbl O, Schmidt NO, Linker R, Hau P, Haedenkamp TM. Effect of antibiotic drug use on outcome and therapy-related toxicity in patients with glioblastoma-A retrospective cohort study. Neurooncol Adv 2024; 6:vdae170. [PMID: 39493414 PMCID: PMC11528512 DOI: 10.1093/noajnl/vdae170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024] Open
Abstract
Background Glioblastoma (GB) is the most frequent malignant brain tumor and has a dismal prognosis. In other cancers, antibiotic use has been associated with severity of chemotherapy-induced toxicity and outcome. We investigated if these mechanisms are also involved in GB. Methods We selected a cohort of 78 GB patients who received combined radiochemotherapy. We investigated if exposure to prediagnostic antibiotic use is associated with clinical side effects and laboratory changes during adjuvant therapy as well as overall survival (OS) and progression-free survival (PFS) using chi-square test, binary logistic regression, Kaplan-Meyer analysis, and multivariable Cox regression. Results Seventeen patients (21.8%) received at least one course of prediagnostic antibiotics and 61 (78.2%) received no antibiotics. We found a higher incidence of loss of appetite (23.5% vs. 4.9%; P = .018) and myelosuppression (41.2% vs. 18.0%; P = .045) in the antibiotic group. Multivariable logistic regression analysis revealed antibiotics to be a predictor for nausea (OR = 6.94, 95% CI: 1.09-44.30; P = .041) and myelosuppression (OR = 9.75, 95% CI: 1.55-61.18; P = .015). Furthermore, lymphocytopenia was more frequent in the antibiotic group (90.0% vs. 56.1%, P = .033). There were no significant differences in OS (P = .404) and PFS (P = .844). Multivariable Cox regression showed a trend toward shorter survival time (P = .089) in the antibiotic group. Conclusions Our study suggests that antibiotic use affects symptoms and lab values in GB patients. Larger prospective studies are required to investigate if prediagnostic antibiotic use could be a prognostic factor in GB patients.
Collapse
Affiliation(s)
- Linda Götz
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany
| | - Tananeh Ansafi
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany
| | - Michael Gerken
- Center for Quality Assurance and Health Services Research, University of Regensburg, Regensburg, Germany
| | - Monika Klinkhammer-Schalke
- Center for Quality Assurance and Health Services Research, University of Regensburg, Regensburg, Germany
| | - Anna Fischl
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany
| | | | - Martin Proescholdt
- Department of Neurosurgery, Regensburg University Hospital, Regensburg, Germany
| | - Elisabeth Bumes
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany
| | - Oliver Kölbl
- Department for Radiotherapy, Regensburg University Hospital, Regensburg, Germany
| | - Nils Ole Schmidt
- Department of Neurosurgery, Regensburg University Hospital, Regensburg, Germany
| | - Ralf Linker
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany
| | - Peter Hau
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany
| | - Tareq M Haedenkamp
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany
| |
Collapse
|
20
|
Almonte AA, Cavic G, Carroll CSE, Neeman T, Fahrer AM. Early T Cell Infiltration Correlates with Anti-CTLA4 Treatment Response in Murine Cancer Models. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1858-1867. [PMID: 37930122 DOI: 10.4049/jimmunol.2300040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 10/12/2023] [Indexed: 11/07/2023]
Abstract
Immune checkpoint inhibitor (ICI) Abs are a revolutionary class of cancer treatment, but only ∼30% of patients receive a lasting benefit from therapy. Preclinical studies using animals from the same genetic backgrounds, challenged with the same cancer models, also show nonuniform responses. Most mouse studies that have evaluated tumor-infiltrating leukocytes after ICI therapy cannot directly correlate their findings with treatment outcomes, because terminal methods were used to acquire immune infiltrate data. In the present study, we used fine-needle aspiration (a nonterminal sampling method) to collect multiple aspirates over several days from s.c. implanted P815, CT26, and 4T1 mouse cancer models treated with ICI Abs. These aspirates were then analyzed with flow cytometry to directly correlate tumor-infiltrating leukocyte populations with treatment success. We found that the P815 and CT26 models respond well to anti-CTLA4 therapies. Among P815-challenged animals, mice that regressed following anti-CTLA4 treatment showed significant increases in CD8+ T cells on days 3, 5, and 7 and in CD4+ T cells on days 5 and 7 and a decrease in macrophages and monocytes on days 3, 5, and 7 after treatment. Similar results were obtained in the CT26 model on day 11 posttreatment. Our study is the first, to our knowledge, to directly correlate early tumor infiltration of T cells with anti-CTLA4 treatment success, thus providing a mechanistic clue toward understanding why alloidentical mice challenged with identical tumors do not respond uniformly to ICI therapies.
Collapse
Affiliation(s)
- Andrew A Almonte
- Division of Biomedical Science and Biochemistry, Research School of Biology, The Australian National University, Canberra, Australia
| | - George Cavic
- Division of Biomedical Science and Biochemistry, Research School of Biology, The Australian National University, Canberra, Australia
| | | | - Teresa Neeman
- Biological Data Science Institute, The Australian National University, Canberra, Australia
| | - Aude M Fahrer
- Division of Biomedical Science and Biochemistry, Research School of Biology, The Australian National University, Canberra, Australia
- Faculty of Science and Technology, University of Canberra, Canberra, Australia
| |
Collapse
|
21
|
Araki T, Kanda S, Ide T, Sonehara K, Komatsu M, Tateishi K, Minagawa T, Kiniwa Y, Kawakami S, Nomura S, Okuyama R, Hanaoka M, Koizumi T. Antiplatelet drugs may increase the risk for checkpoint inhibitor-related pneumonitis in advanced cancer patients. ESMO Open 2023; 8:102030. [PMID: 37852033 PMCID: PMC10774871 DOI: 10.1016/j.esmoop.2023.102030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/31/2023] [Accepted: 09/20/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are indicated for various cancers and are the mainstay of cancer immunotherapy. They are often associated with ICI-related pneumonitis (CIP), however, hindering a favorable clinical course. Recently, non-oncology concomitant drugs have been reported to affect the efficacy and toxicity of ICIs; however, the association between these drugs and the risk for CIP is uncertain. The aim of this study was to assess the impact of baseline concomitant drugs on CIP incidence in ICI-treated advanced cancer patients. PATIENTS AND METHODS This was a single-center retrospective study that included a cohort of 511 patients with advanced cancer (melanoma and non-small-cell lung, head and neck, genitourinary, and other types of cancer) treated with ICIs. Univariable analysis was conducted to identify baseline co-medications associated with CIP incidence. A propensity score matching analysis was used to adjust for potential CIP risk factors, and multivariable analysis was carried out to assess the impact of the identified co-medications on CIP risk. RESULTS Forty-seven (9.2%) patients developed CIP. In these patients, the organizing pneumonia pattern was the dominant radiological phenotype, and 42.6% had grade ≥3 CIP, including one patient with grade 5. Of the investigated baseline co-medications, the proportion of antiplatelet drugs (n = 50, 9.8%) was higher in patients with CIP (23.4% versus 8.4%). After propensity score matching, the CIP incidence was higher in patients with baseline antiplatelet drugs (22% versus 6%). Finally, baseline antiplatelet drug use was demonstrated to increase the risk for CIP incidence regardless of cancer type (hazard ratio, 3.46; 95% confidence interval 1.21-9.86). CONCLUSIONS An association between concomitant antiplatelet drug use at baseline and an increased risk for CIP was seen in our database. This implies the importance of assessing concomitant medications for CIP risk management.
Collapse
Affiliation(s)
- T Araki
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - S Kanda
- Department of Hematology and Medical Oncology, Shinshu University School of Medicine, Matsumoto, Japan.
| | - T Ide
- Department of Pharmacy, Shinshu University School of Medicine, Matsumoto, Japan
| | - K Sonehara
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - M Komatsu
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - K Tateishi
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - T Minagawa
- Department of Urology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Y Kiniwa
- Department of Dermatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - S Kawakami
- Department of Radiology, Shinshu University School of Medicine, Matsumoto, Japan
| | - S Nomura
- Department of Biostatistics and Bioinformatics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - R Okuyama
- Department of Dermatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - M Hanaoka
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - T Koizumi
- Department of Hematology and Medical Oncology, Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
22
|
Dharmapuri S, Özbek U, Jethra H, Jun T, Marron TU, Saeed A, Huang YH, Muzaffar M, Pinter M, Balcar L, Fulgenzi C, Amara S, Weinmann A, Personeni N, Scheiner B, Pressiani T, Navaid M, Bengsch B, Paul S, Khan U, Bettinger D, Nishida N, Mohamed YI, Vogel A, Gampa A, Korolewicz J, Cammarota A, Kaseb A, Galle PR, Pillai A, Wang YH, Cortellini A, Kudo M, D’Alessio A, Rimassa L, Pinato DJ, Ang C. Baseline neutrophil-lymphocyte ratio and platelet-lymphocyte ratio appear predictive of immune treatment related toxicity in hepatocellular carcinoma. World J Gastrointest Oncol 2023; 15:1900-1912. [PMID: 38077640 PMCID: PMC10701235 DOI: 10.4251/wjgo.v15.i11.1900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 09/14/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND A well-recognized class effect of immune checkpoint inhibitors (ICI) is immune-related adverse events (IrAEs) ranging from low grade toxicities to life-threatening end organ damage requiring permanent discontinuation of ICI. Deaths are reported in < 5% of patients treated with ICI. There are, however, no reliable markers to predict the onset and severity of IrAEs. We tested the association between neutrophil-lymphocyte ratio (NLR) and platelet-lymphocyte ratio (PLR) at baseline with development of clinically significant IrAEs (grade ≥ 2) in hepatocellular carcinoma (HCC) patients treated with ICI. AIM To test the association between NLR and PLR at baseline with development of clinically significant IrAEs (grade ≥ 2) in HCC patients treated with ICI. METHODS Data was extracted from an international database from a consortium of 11 tertiary-care referral centers. NLR = absolute neutrophil count/absolute lymphocyte count (ALC) and PLR = platelet count/ALC. Cutoff of 5 was used for NLR and 300 for PLR based on literature. We also tested the association between antibiotic and steroid exposure to IrAEs. RESULTS Data was collected from 361 patients treated between 2016-2020 across the United States (67%), Asia (14%) and Europe (19%). Most patients received Nivolumab (n = 255, 71%). One hundred sixty-seven (46%) patients developed at least one IrAE, highest grade 1 in 80 (48%), grade ≥ 2 in 87 (52%) patients. In a univariable regression model PLR > 300 was significantly associated with a lower incidence of grade ≥ 2 IrAEs (OR = 0.40; P = 0.044). Similarly, a trend was observed between NLR > 5 and lower incidence of grade ≥ 2 IrAEs (OR = 0.58; P = 0.097). Multivariate analyses confirmed PLR > 300 as an independent predictive marker of grade ≥ 2 IrAEs (OR = 0.26; P = 0.011), in addition to treatment with programmed cell death ligand 1 (PD-1)/cytotoxic T lymphocyte-associated protein-4 (OR = 2.57; P = 0.037) and PD-1/tyrosine kinase inhibitor (OR = 3.39; P = 0.01) combinations. Antibiotic use was not associated with IrAE incidence (OR = 1.02; P = 0.954). Patients treated with steroids had a > 2-fold higher incidence of grade ≥ 2 IrAEs (OR = 2.74; P < 0.001), although 74% were prescribed steroids for the treatment of IrAEs. CONCLUSION Given that high baseline NLR and PLR are associated with a decreased incidence of IrAEs, lower baseline NLR and PLR may be predictive biomarkers for the appearance of IrAEs in HCC treated with ICI. This finding is in keeping with several studies in solid tumors that have shown that baseline NLR and PLR appear predictive of IrAEs.
Collapse
Affiliation(s)
- Sirish Dharmapuri
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Umut Özbek
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Hiren Jethra
- Department of Data Analytics Harrisburg, Harrisburg University of Science and Technology, Harrisburd, PA 17101, United States
| | - Tomi Jun
- SEMA4, Stamford, CT 06902, United States
| | - Thomas U Marron
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Anwaar Saeed
- Division of Medical Oncology Kansas, University of Kansas Cancer Center, Kansas, MO 66160, United States
| | - Yi-Hsiang Huang
- Division of Gastroenterology and Hepatology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Mahvish Muzaffar
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27858, United States
| | - Matthias Pinter
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna 1090, Austria
| | - Lorenz Balcar
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna 1090, Austria
| | - Claudia Fulgenzi
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital London, London W12 0HS, United Kingdom
| | - Suneetha Amara
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27858, United States
| | - Arndt Weinmann
- Department of Hepatology, Johannes Gutenberg-University Medical Centre, Niedersachsen 30625, Germany
| | - Nicola Personeni
- Medical Oncology Unit, ASST Garda, Via Lungomella Valsecchi, Brescia, Manerbio 25025, Italy
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Milan, Rozzano 20089, Italy
| | - Bernhard Scheiner
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna 1090, Austria
| | - Tiziana Pressiani
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Milan, Rozzano 20089, Italy
| | - Musharraf Navaid
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27858, United States
| | - Bertram Bengsch
- Department of Medicine II, Univ Med Ctr Freiburg, Hugstetter Str 55, University Hospital Freiburg, Freiburg D-79106, Germany
| | - Sonal Paul
- Department of Oncology Baltimore, LifeBridge Health, Baltimore, MD 21215, United States
| | - Uqba Khan
- Division of Hematology and Oncology, Weill Cornell Medical College, NY 10065, United States
| | - Dominik Bettinger
- Department of Medicine II, Univ Med Ctr Freiburg, Hugstetter Str 55, University Hospital Freiburg, Freiburg D-79106, Germany
| | - Naoshi Nishida
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka 577-8502, Japan
| | - Yehia Ibrahim Mohamed
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Arndt Vogel
- Department of Gastroenterology Hepatology and Endocrinology, HannoverArndt Vogel, Medical School Hannover, Carl-Neubergstr., Hannover 30659, Germany
| | - Anuhya Gampa
- Department of Hepatology, Rush University Medical Group 1725 W Harrison St Ste 158, Chicago, IL 60612, United States
| | - James Korolewicz
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital London, London W12 0HS, United Kingdom
| | - Antonella Cammarota
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Milan, Rozzano 20089, Italy
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Milan, Pieve Emanuele 20072, Italy
| | - Ahmed Kaseb
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Peter R Galle
- Department of Internal Medicine I and Cirrhosis Center Mainz, University Medical Center Mainz, Johannes Gutenberg Univ Mainz, Med Klin and Poliklin, Mainz D-55131, Germany
| | - Anjana Pillai
- Department of Gastroenterology, Hepatology, and Nutrition, University of Chicago Medical Center, Chicago, IL 60637, United States
| | - Ying-Hong Wang
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Alessio Cortellini
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital London, London W12 0HS, United Kingdom
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka 577-8502, Japan
| | - Antonio D’Alessio
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital London, London W12 0HS, United Kingdom
| | - Lorenza Rimassa
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Milan, Rozzano 20089, Italy
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Milan, Pieve Emanuele 20072, Italy
| | - David James Pinato
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital London, London W12 0HS, United Kingdom
| | - Celina Ang
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| |
Collapse
|
23
|
Guillot N, Roméo B, Manesh SS, Milano G, Brest P, Zitvogel L, Hofman P, Mograbi B. Manipulating the gut and tumor microbiota for immune checkpoint inhibitor therapy: from dream to reality. Trends Mol Med 2023; 29:897-911. [PMID: 37704493 DOI: 10.1016/j.molmed.2023.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 09/15/2023]
Abstract
The past decade has witnessed a revolution in cancer treatment by shifting from conventional therapies to immune checkpoint inhibitors (ICIs). These immunotherapies unleash the host immune system against the tumor and have achieved unprecedented durable remission. However, 80% of patients do not respond. This review discusses how bacteria are unexpected drivers that reprogram tumor immunity. Manipulating the microbiota impacts on tumor development and reprograms the tumor microenvironment (TME) of mice on immunotherapy. We anticipate that harnessing commensals and the tumor microbiome holds promise to identify patients who will benefit from immunotherapy and guide the choice of new ICI combinations to advance treatment efficacy.
Collapse
Affiliation(s)
- Nicolas Guillot
- Université Côte d'Azur, CNRS, INSERM, IRCAN, IHU RespirERA, FHU OncoAge, Centre Antoine Lacassagne, Nice, France
| | - Barnabé Roméo
- Université Côte d'Azur, CNRS, INSERM, IRCAN, IHU RespirERA, FHU OncoAge, Centre Antoine Lacassagne, Nice, France
| | - Shima Sepehri Manesh
- Université Côte d'Azur, CNRS, INSERM, IRCAN, IHU RespirERA, FHU OncoAge, Centre Antoine Lacassagne, Nice, France
| | - Gerard Milano
- Centre Antoine Lacassagne, Service de Valorisation Scientifique, Nice, France
| | - Patrick Brest
- Université Côte d'Azur, CNRS, INSERM, IRCAN, IHU RespirERA, FHU OncoAge, Centre Antoine Lacassagne, Nice, France
| | - Laurence Zitvogel
- Gustave Roussy, Villejuif, France; Equipe Labellisée par la Ligue Contre le Cancer, INSERM Unité 1015, Villejuif, France; Université Paris Saclay, Faculty of Medicine, Le Kremlin-Bicêtre, France
| | - Paul Hofman
- Université Côte d'Azur, CNRS, INSERM, IRCAN, IHU RespirERA, FHU OncoAge, Centre Antoine Lacassagne, Nice, France; University Côte d'Azur, IHU RespirERA, FHU OncoAge, CHU of Nice, Laboratory of Clinical and Experimental Pathology (LPCE), Biobank (BB-0033-00025), Nice, France
| | - Baharia Mograbi
- Université Côte d'Azur, CNRS, INSERM, IRCAN, IHU RespirERA, FHU OncoAge, Centre Antoine Lacassagne, Nice, France.
| |
Collapse
|
24
|
Guo C, Kong L, Xiao L, Liu K, Cui H, Xin Q, Gu X, Jiang C, Wu J. The impact of the gut microbiome on tumor immunotherapy: from mechanism to application strategies. Cell Biosci 2023; 13:188. [PMID: 37828613 PMCID: PMC10571290 DOI: 10.1186/s13578-023-01135-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 09/15/2023] [Indexed: 10/14/2023] Open
Abstract
Immunotherapy is one of the fastest developing areas in the field of oncology. Many immunological treatment strategies for refractory tumors have been approved and marketed. Nevertheless, much clinical and preclinical experimental evidence has shown that the efficacy of immunotherapy in tumor treatment varies markedly among individuals. The commensal microbiome mainly colonizes the intestinal lumen in humans, is affected by a variety of factors and exhibits individual variation. Moreover, the gut is considered the largest immune organ of the body due to its influence on the immune system. In the last few decades, with the development of next-generation sequencing (NGS) techniques and in-depth research, the view that the gut microbiota intervenes in antitumor immunotherapy through the immune system has been gradually confirmed. Here, we review important studies published in recent years focusing on the influences of microbiota on immune system and the progression of malignancy. Furthermore, we discuss the mechanism by which microbiota affect tumor immunotherapy, including immune checkpoint blockade (ICB) and adoptive T-cell therapy (ACT), and strategies for modulating the microbial composition to facilitate the antitumor immune response. Finally, opportunity and some challenges are mentioned to enable a more systematic understanding of tumor treatment in the future and promote basic research and clinical application in related fields.
Collapse
Affiliation(s)
- Ciliang Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, 22 Hankou Road, Nanjing, 210093, Jiangsu, China
| | - Lingkai Kong
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, 22 Hankou Road, Nanjing, 210093, Jiangsu, China
| | - Lingjun Xiao
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, 22 Hankou Road, Nanjing, 210093, Jiangsu, China
| | - Kua Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, 22 Hankou Road, Nanjing, 210093, Jiangsu, China
| | - Huawei Cui
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, 22 Hankou Road, Nanjing, 210093, Jiangsu, China
| | - Qilei Xin
- Jinan Microecological Biomedicine Shandong Laboratory, Shounuo City Light West Block, Qingdao Road 3716#, Huaiyin District, Jinan, Shandong, China
| | - Xiaosong Gu
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, 22 Hankou Road, Nanjing, 210093, Jiangsu, China
- Jinan Microecological Biomedicine Shandong Laboratory, Shounuo City Light West Block, Qingdao Road 3716#, Huaiyin District, Jinan, Shandong, China
| | - Chunping Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, 22 Hankou Road, Nanjing, 210093, Jiangsu, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Shounuo City Light West Block, Qingdao Road 3716#, Huaiyin District, Jinan, Shandong, China.
| | - Junhua Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, 22 Hankou Road, Nanjing, 210093, Jiangsu, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Shounuo City Light West Block, Qingdao Road 3716#, Huaiyin District, Jinan, Shandong, China.
| |
Collapse
|
25
|
Allais BS, Fay CJ, Kim DY, Semenov YR, LeBoeuf NR. Cutaneous immune-related adverse events from immune checkpoint inhibitor therapy: Moving beyond "maculopapular rash". Immunol Rev 2023; 318:22-36. [PMID: 37583051 DOI: 10.1111/imr.13257] [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: 04/05/2023] [Accepted: 06/27/2023] [Indexed: 08/17/2023]
Abstract
Uncoupling toxicity from therapeutic effect lies at the foundation of the current state of the field of cutaneous immune-related adverse events to immune checkpoint inhibitor therapy. This will be achieved through understanding the drivers of toxicity, tumor response, and resistance via large, well-powered population-level studies, institutional cohort data, and cellular-level data. Increasing diagnostic specificity through the application of consensus disease definitions has the power to improve clinical care and each approach to research. Cutaneous immune-related adverse events are associated with increased survival, and their treatment must invoke the maintenance of a delicate balance between immunosuppression, anti-tumor effect of immune checkpoint inhibitor therapy, and quality of life. The multidisciplinary care of cancer patients with adverse events is critical to optimizing clinical and translational research outcomes and, as such, dermatologists are vital to moving the study of cutaneous adverse events forward.
Collapse
Affiliation(s)
- Blair S Allais
- Inova Schar Cancer Institute, Melanoma and Skin Cancer Center, Fairfax, Virginia, USA
| | - Christopher J Fay
- The Center for Cutaneous Oncology, Department of Dermatology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Daniel Y Kim
- Harvard-MIT Health Sciences and Technology Program, Harvard Medical School, Boston, Massachusetts, USA
| | - Yevgeniy R Semenov
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Nicole R LeBoeuf
- The Center for Cutaneous Oncology, Department of Dermatology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Fortman DD, Hurd D, Davar D. The Microbiome in Advanced Melanoma: Where Are We Now? Curr Oncol Rep 2023; 25:997-1016. [PMID: 37269504 PMCID: PMC11090495 DOI: 10.1007/s11912-023-01431-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2023] [Indexed: 06/05/2023]
Abstract
PURPOSE OF REVIEW This review summarizes recent data linking gut microbiota composition to ICI outcomes and gut microbiota-specific interventional clinical trials in melanoma. RECENT FINDINGS Preclinical and clinical studies have demonstrated the effects of the gut microbiome modulation upon ICI response in advanced melanoma, with growing evidence supporting the ability of the gut microbiome to restore or improve ICI response in advanced melanoma through dietary fiber, probiotics, and FMT. Immune checkpoint inhibitors (ICI) targeting the PD-1, CTLA-4, and LAG-3 negative regulatory checkpoints have transformed the management of melanoma. ICIs are FDA-approved in advanced metastatic disease, stage III resected melanoma, and high-risk stage II melanoma and are being investigated more recently in the management of high-risk resectable melanoma in the peri-operative setting. The gut microbiome has emerged as an important tumor-extrinsic modulator of both response and immune-related adverse event (irAE) development in ICI-treated cancer in general, and melanoma in particular.
Collapse
Affiliation(s)
- Dylan D Fortman
- Division of General Internal Medicine, Department of Medicine, University of Pittsburgh Medical Center (UPMC) and University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Drew Hurd
- UPMC Hillman Cancer Center, Department of Medicine, University of Pittsburgh, Pavilion, Suite 1.32d, 5115, Center Avenue, Pittsburgh, PA, 15213, USA
| | - Diwakar Davar
- UPMC Hillman Cancer Center, Department of Medicine, University of Pittsburgh, Pavilion, Suite 1.32d, 5115, Center Avenue, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
27
|
Li L, Tianrui K, Chunlei L, Zhendong Q, Xiaoyan C, Wenhong D. HYDIN mutation status as a potential predictor of immune checkpoint inhibitor efficacy in melanoma. Aging (Albany NY) 2023; 15:7997-8012. [PMID: 37595251 PMCID: PMC10496993 DOI: 10.18632/aging.204925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/10/2023] [Indexed: 08/20/2023]
Abstract
BACKGROUND The advent of immune checkpoint inhibitors (ICIs) has altered the outlook for cancer treatment. The estimation of predictive biomarkers could contribute to maximizing the benefits from ICIs treatment. Here, we explored the association between HYDIN mutations (HYDIN-MUT) in melanoma and ICIs efficacy. METHODS Clinical data and sequencing data from published studies were utilized to assess the association between HYDIN-MUT and the efficacy of ICIs treatment in melanoma patients. RESULTS Compared to other tumor types, HYDIN (36.14%) has the highest mutation rate in melanoma patients. In the anti-PD-1 treated cohort (n = 254), the HYDIN-MUT patients had a longer OS after ICIs treatment than the HYDIN wild-type (HYDIN-WT) patients (HR = 0.590 [95% CI, 0.410-0.847], P = 0.004); the objective response rate (ORR) and durable clinical benefit (DCB) were increased in patients with HYDIN-MUT (ORR = 46.25, DCB = 56.00%) compared to patients with HYDIN-WT (ORR = 30.99%, DCB = 42.76%) (ORR: P = 0.019; DCB: P = 0.060). In the anti-CTLA4 treated cohort (n = 174), HYDIN-MUT patients achieved significantly longer OS than HYDIN-WT patients (HR = 0.549 [95% CI, 0.366-0.823], P = 0.003); the proportion of ORR and DCB in HYDIN-MUT patients was significantly higher than that in HYDIN-WT patients (ORR 40.54% vs. 14.42%, P = 0.031; DCB 45.76% vs. 22.22%, P = 0.002). Further gene set enrichment analysis demonstrated that DNA repair and anti-tumor immunity were significantly enhanced in HYDIN-MUT patients. CONCLUSIONS HYDIN mutations are a potential predictive biomarker of ICIs efficacy in melanoma patients.
Collapse
Affiliation(s)
- Liu Li
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Kuang Tianrui
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Li Chunlei
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Qiu Zhendong
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Chen Xiaoyan
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Deng Wenhong
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
28
|
Chen N, Xu X, Fan Y. Immune checkpoint inhibitors in the treatment of oesophageal squamous cell carcinoma: where are we and where are we going? Ther Adv Med Oncol 2023; 15:17588359231189420. [PMID: 37547447 PMCID: PMC10399266 DOI: 10.1177/17588359231189420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 07/05/2023] [Indexed: 08/08/2023] Open
Abstract
Oesophageal squamous cell carcinoma (ESCC) is a kind of malignant tumour with high invasiveness and a poor prognosis. Immunotherapy, especially immune checkpoint inhibitors (ICIs), is a rapidly growing therapeutic method that activates and enhances anti-tumour immunity to treat patients with malignancy. Several clinical trials have confirmed the efficacy of ICIs in the treatment of ESCC. ICIs have been approved for the treatment of patients with ESCC. However, only a subset of patients can obtain excellent benefits from ICI therapy. In recent years, there has been a growing interest in exploring predictive biomarkers of immunotherapy response. In this review, we highlighted the predictive biomarkers for the prognosis of ESCC patients treated with ICIs and pointed out the existing problems and the direction of future research in this field.
Collapse
Affiliation(s)
- Ning Chen
- Department of Oncology, The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xiaoling Xu
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, No. 1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang 310022, China
| | - Yun Fan
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, No. 1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
29
|
Pizzutilo EG, Romanò R, Roazzi L, Agostara AG, Oresti S, Zeppellini A, Giannetta L, Cerea G, Signorelli D, Siena S, Sartore-Bianchi A. Immune Checkpoint Inhibitors and the Exposome: Host-Extrinsic Factors Determine Response, Survival, and Toxicity. Cancer Res 2023; 83:2283-2296. [PMID: 37205627 PMCID: PMC10345966 DOI: 10.1158/0008-5472.can-23-0161] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/24/2023] [Accepted: 05/15/2023] [Indexed: 05/21/2023]
Abstract
Cancer immunotherapy, largely represented by immune checkpoint inhibitors (ICI), has led to substantial changes in preclinical cancer research and clinical oncology practice over the past decade. However, the efficacy and toxicity profiles of ICIs remain highly variable among patients, with only a fraction achieving a significant benefit. New combination therapeutic strategies are being investigated, and the search for novel predictive biomarkers is ongoing, mainly focusing on tumor- and host-intrinsic components. Less attention has been directed to all the external, potentially modifiable factors that compose the exposome, including diet and lifestyle, infections, vaccinations, and concomitant medications, that could affect the immune system response and its activity against cancer cells. We hereby provide a review of the available clinical evidence elucidating the impact of host-extrinsic factors on ICI response and toxicity.
Collapse
Affiliation(s)
- Elio Gregory Pizzutilo
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Università degli Studi di Milano, Department of Oncology and Hemato-Oncology, Milan, Italy
| | - Rebecca Romanò
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Università degli Studi di Milano, Department of Oncology and Hemato-Oncology, Milan, Italy
| | - Laura Roazzi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Università degli Studi di Milano, Department of Oncology and Hemato-Oncology, Milan, Italy
| | - Alberto G. Agostara
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Università degli Studi di Milano, Department of Oncology and Hemato-Oncology, Milan, Italy
| | - Sara Oresti
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Università degli Studi di Milano, Department of Oncology and Hemato-Oncology, Milan, Italy
| | - Annalisa Zeppellini
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Laura Giannetta
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giulio Cerea
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Diego Signorelli
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Università degli Studi di Milano, Department of Oncology and Hemato-Oncology, Milan, Italy
| | - Andrea Sartore-Bianchi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Università degli Studi di Milano, Department of Oncology and Hemato-Oncology, Milan, Italy
| |
Collapse
|
30
|
Curkovic NB, Johnson DB. Updates in toxicities associated with immune checkpoint inhibitors. Expert Rev Clin Immunol 2023; 19:1117-1129. [PMID: 37276071 PMCID: PMC10527235 DOI: 10.1080/1744666x.2023.2221434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/31/2023] [Indexed: 06/07/2023]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) have become a pillar of treatment for numerous cancers with increasing use in combination with other ICIs and in earlier stages of disease treatment. Although effective, ICI use is accompanied by a milieu of potentially bothersome or even life-threatening toxicities known as immune-related adverse events (irAEs), necessitating careful monitoring and early intervention. AREAS COVERED In this review, we provide an overview of recent advances surrounding toxicity pathophysiology and treatment in the context of relevant organ systems. An emphasis on current treatments by toxicity, as well as updates on steroid-refractory toxicities, chronic toxicities, and biomarkers will be a focus of this update on the current understanding of irAEs. EXPERT OPINION ICI toxicities are a major limitation on the deployment of multi-agent ICI regimens and are thus a major priority to understand, treat, and prevent. Recent developments have led to greater understanding of the pathophysiology of these events, which may lead to improved prevention or mitigation strategies. Further, early studies have also suggested steroid-sparing approaches that may be useful. Ultimately, preventing and managing irAEs will be a key goal toward successful ICI treatment across a broader range of patients with cancer.
Collapse
Affiliation(s)
| | - Douglas B. Johnson
- Department of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
31
|
Fidelle M, Rauber C, Alves Costa Silva C, Tian AL, Lahmar I, de La Varende ALM, Zhao L, Thelemaque C, Lebhar I, Messaoudene M, Pizzato E, Birebent R, Mbogning Fonkou MD, Zoppi S, Reni A, Dalban C, Leduc M, Ferrere G, Durand S, Ly P, Silvin A, Mulder K, Dutertre CA, Ginhoux F, Yonekura S, Roberti MP, Tidjani-Alou M, Terrisse S, Chen J, Kepp O, Schippers A, Wagner N, Suárez-Gosálvez J, Kobold S, Fahrner JE, Richard C, Bosq J, Lordello L, Vitali G, Galleron N, Quinquis B, Le Chatelier E, Blanchard L, Girard JP, Jarry A, Gervois N, Godefroy E, Labarrière N, Koschny R, Daillère R, Besse B, Truntzer C, Ghiringhelli F, Coatnoan N, Mhanna V, Klatzmann D, Drubay D, Albiges L, Thomas AM, Segata N, Danlos FX, Marabelle A, Routy B, Derosa L, Kroemer G, Zitvogel L. A microbiota-modulated checkpoint directs immunosuppressive intestinal T cells into cancers. Science 2023; 380:eabo2296. [PMID: 37289890 DOI: 10.1126/science.abo2296] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/14/2023] [Indexed: 06/10/2023]
Abstract
Antibiotics (ABX) compromise the efficacy of programmed cell death protein 1 (PD-1) blockade in cancer patients, but the mechanisms underlying their immunosuppressive effects remain unknown. By inducing the down-regulation of mucosal addressin cell adhesion molecule 1 (MAdCAM-1) in the ileum, post-ABX gut recolonization by Enterocloster species drove the emigration of enterotropic α4β7+CD4+ regulatory T 17 cells into the tumor. These deleterious ABX effects were mimicked by oral gavage of Enterocloster species, by genetic deficiency, or by antibody-mediated neutralization of MAdCAM-1 and its receptor, α4β7 integrin. By contrast, fecal microbiota transplantation or interleukin-17A neutralization prevented ABX-induced immunosuppression. In independent lung, kidney, and bladder cancer patient cohorts, low serum levels of soluble MAdCAM-1 had a negative prognostic impact. Thus, the MAdCAM-1-α4β7 axis constitutes an actionable gut immune checkpoint in cancer immunosurveillance.
Collapse
Affiliation(s)
- Marine Fidelle
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Conrad Rauber
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Department of Gastroenterology and Infectious Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Carolina Alves Costa Silva
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Ai-Ling Tian
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Imran Lahmar
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Anne-Laure Mallard de La Varende
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Liwei Zhao
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Cassandra Thelemaque
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Isabelle Lebhar
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Meriem Messaoudene
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada
| | - Eugenie Pizzato
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Roxanne Birebent
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Maxime Descartes Mbogning Fonkou
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Silvia Zoppi
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Anna Reni
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Cécile Dalban
- Clinical Research Department, Centre Léon Bérard, Lyon, France
| | - Marion Leduc
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Gladys Ferrere
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- EverImmune, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Sylvère Durand
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Pierre Ly
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS), Villejuif, France
| | - Aymeric Silvin
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Kevin Mulder
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Charles-Antoine Dutertre
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Florent Ginhoux
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Satoru Yonekura
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Maria Paula Roberti
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD), Heidelberg, Germany
| | - Maryam Tidjani-Alou
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Safae Terrisse
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Jianzhou Chen
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Oliver Kepp
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Angela Schippers
- Department of Pediatrics, University Hospital RWTH Aachen, Aachen, Germany
| | - Norbert Wagner
- Department of Pediatrics, University Hospital RWTH Aachen, Aachen, Germany
| | - Javier Suárez-Gosálvez
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| | - Jean-Eudes Fahrner
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Corentin Richard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada
| | | | - Leonardo Lordello
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Giacomo Vitali
- MetaGenoPolis, INRAe, Université Paris-Saclay, Jouy en Josas, France
| | - Nathalie Galleron
- MetaGenoPolis, INRAe, Université Paris-Saclay, Jouy en Josas, France
| | - Benoît Quinquis
- MetaGenoPolis, INRAe, Université Paris-Saclay, Jouy en Josas, France
| | | | - Lucas Blanchard
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Jean-Philippe Girard
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Anne Jarry
- Nantes Université, Université d'Angers, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302/EMR6001, Nantes, France
| | - Nadine Gervois
- Nantes Université, Université d'Angers, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302/EMR6001, Nantes, France
| | - Emmanuelle Godefroy
- Nantes Université, Université d'Angers, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302/EMR6001, Nantes, France
| | - Nathalie Labarrière
- Nantes Université, Université d'Angers, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302/EMR6001, Nantes, France
- LabEx IGO, Université de Nantes, Nantes, France
| | - Ronald Koschny
- Department of Gastroenterology and Infectious Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Romain Daillère
- EverImmune, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Benjamin Besse
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
| | - Caroline Truntzer
- Université de Bourgogne Franche-Comté, Plateforme de Transfert de Biologie du Cancer, Centre Georges-François Leclerc, Equipe Labellisée Ligue Nationale Contre le Cancer, Centre de Recherche INSERM LNC-UMR1231, Institut Médical de Génétique et d'Immunologie, Dijon, France
| | - François Ghiringhelli
- Université de Bourgogne Franche-Comté, Plateforme de Transfert de Biologie du Cancer, Centre Georges-François Leclerc, Equipe Labellisée Ligue Nationale Contre le Cancer, Centre de Recherche INSERM LNC-UMR1231, Institut Médical de Génétique et d'Immunologie, Dijon, France
| | - Nicolas Coatnoan
- AP-HP, Hôpital Pitié-Salpêtrière, Clinical Investigation Center for Biotherapies (CIC-BTi) and Immunology-Inflammation-Infectiology and Dermatology Department (3iD), Paris, France
- Sorbonne Université, INSERM, UMRS959 Immunology-Immunopathology-Immunotherapy Laboratory, Paris, France
| | - Vanessa Mhanna
- AP-HP, Hôpital Pitié-Salpêtrière, Clinical Investigation Center for Biotherapies (CIC-BTi) and Immunology-Inflammation-Infectiology and Dermatology Department (3iD), Paris, France
- Sorbonne Université, INSERM, UMRS959 Immunology-Immunopathology-Immunotherapy Laboratory, Paris, France
| | - David Klatzmann
- AP-HP, Hôpital Pitié-Salpêtrière, Clinical Investigation Center for Biotherapies (CIC-BTi) and Immunology-Inflammation-Infectiology and Dermatology Department (3iD), Paris, France
- Sorbonne Université, INSERM, UMRS959 Immunology-Immunopathology-Immunotherapy Laboratory, Paris, France
| | - Damien Drubay
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Office of Biostatistics and Epidemiology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
- Inserm, Université Paris-Saclay, CESP U1018, Oncostat, labeled Ligue Contre le Cancer, Villejuif, France
| | - Laurence Albiges
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
| | - Andrew Maltez Thomas
- Department of Computational, Cellular and Integrative Biology, University of Trento, Trento, Italy
| | - Nicola Segata
- Department of Computational, Cellular and Integrative Biology, University of Trento, Trento, Italy
- Istituto Europeo di Oncologia (IEO), National Cancer Institute (IRCCS), Milan, Italy
| | - François-Xavier Danlos
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS), Villejuif, France
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Aurélien Marabelle
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS), Villejuif, France
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Bertrand Routy
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada
- Hematology-Oncology Division, Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Quebec, Canada
| | - Lisa Derosa
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS), Villejuif, France
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS), Villejuif, France
| |
Collapse
|
32
|
Golčić M, Simetić L, Herceg D, Blažičević K, Kenđel Jovanović G, Dražić I, Belančić A, Skočibušić N, Palčevski D, Rubinić I, Vlahović-Palčevski V, Majnarić T, Dobrila-Dintinjana R, Pleština S. Analysis of the Gut Microbiome and Dietary Habits in Metastatic Melanoma Patients with a Complete and Sustained Response to Immunotherapy. Cancers (Basel) 2023; 15:cancers15113052. [PMID: 37297014 DOI: 10.3390/cancers15113052] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/26/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Immunotherapy has improved the prognosis of metastatic melanoma patients, although most patients do not achieve a complete response. While specific gut microbiome and dietary habits might influence treatment success, there is a lack of concordance between the studies, potentially due to dichotomizing patients only into responders and non-responders. The aim of this study was to elucidate whether metastatic melanoma patients with complete and sustained response to immunotherapy exhibit differences in gut microbiome composition among themselves, and whether those differences were associated with specific dietary habits. Shotgun metagenomic sequencing revealed that patients who exhibited a complete response after more than 9 months of treatment (late responders) exhibited a significantly higher beta-diversity (p = 0.02), with a higher abundance of Coprococcus comes (LDA 3.548, p = 0.010), Bifidobacterium pseudocatenulatum (LDA 3.392, p = 0.024), and lower abundance of Prevotellaceae (p = 0.04) compared to early responders. Furthermore, late responders exhibited a different diet profile, with a significantly lower intake of proteins and sweets and a higher intake of flavones (p < 0.05). The research showed that metastatic melanoma patients with a complete and sustained response to immunotherapy were a heterogeneous group. Patients with a late complete response exhibited microbiome and dietary habits which were previously associated with an improved response to immunotherapy.
Collapse
Affiliation(s)
- Marin Golčić
- Department of Radiotherapy and Oncology, University Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Luka Simetić
- Department of Oncology, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| | - Davorin Herceg
- Department of Oncology, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| | - Krešimir Blažičević
- Department of Oncology, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| | - Gordana Kenđel Jovanović
- Department of Health Ecology, Teaching Institute of Public Health of Primorsko-Goranska County, 51000 Rijeka, Croatia
| | - Ivan Dražić
- Department of Mathematics, Physics and Foreign Languages, Faculty of Engineering, University of Rijeka, 51000 Rijeka, Croatia
| | - Andrej Belančić
- Department of Clinical Pharmacology, University Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Nataša Skočibušić
- Department of Clinical Pharmacology, University Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Dora Palčevski
- Department of Internal Medicine, University Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Igor Rubinić
- Department of Clinical Pharmacology, University Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Vera Vlahović-Palčevski
- Department of Clinical Pharmacology, University Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Tea Majnarić
- Community Health Center of Primorsko-Goranska County, 51000 Rijeka, Croatia
| | | | - Stjepko Pleština
- Department of Oncology, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
33
|
Argoubi R, Reese ES, Furegato M, Medina P, Bobiak S. Advanced or metastatic biliary tract cancer in Japan: a study using the Japan Medical Data Center payer claims database. J Comp Eff Res 2023; 12:e220201. [PMID: 37256267 PMCID: PMC10402906 DOI: 10.57264/cer-2022-0201] [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: 12/02/2022] [Accepted: 05/02/2023] [Indexed: 06/01/2023] Open
Abstract
Aim: Biliary tract cancers are aggressive, with poor prognosis. This study describes clinical characteristics, treatment patterns and healthcare resource utilization in patients with metastatic biliary tract cancer in Japan. Materials & methods: This cohort-based study collected data from the Japan Medical Data Center claims database (2014-2018). Results: A total of 325 patients were included; 65.2% were male and the mean age was 59.2 years. A 47.6% had an Elixhauser Comorbidity Index score ≥5. Most frequent regimens were gemcitabine + cisplatin (52.9%) for first-line therapy and tegafur + gimeracil + oteracil for second-line therapy (48.6%) and third-line therapy (27.2%). Approximately 77% of patients had ≥1 hospital admission, with a median length of 57 days. Conclusion: This study provides insights on the characteristics and burden of metastatic biliary tract cancer in Japan, highlighting high disease burden in a younger population.
Collapse
Affiliation(s)
| | - Emily S Reese
- EMD Serono Research & Development Institute, Inc, Billerica, MA 01821, USA, an affiliate of Merck KGaA
| | | | | | - Sarah Bobiak
- EMD Serono Research & Development Institute, Inc, Billerica, MA 01821, USA, an affiliate of Merck KGaA
| |
Collapse
|
34
|
Delaye M, Rousseau A, Mailly-Giacchetti L, Assoun S, Sokol H, Neuzillet C. Obesity, cancer, and response to immune checkpoint inhibitors: Could the gut microbiota be the mechanistic link? Pharmacol Ther 2023:108442. [PMID: 37210004 DOI: 10.1016/j.pharmthera.2023.108442] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/27/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Immune checkpoint inhibitors (ICI) have deeply changed the therapeutic management of a broad spectrum of solid tumors. Recent observations showed that obese patients receiving ICIs might have better outcomes than those with normal weight, while obesity was historically associated with a worse prognosis in cancer patients. Of note, obesity is associated with alterations in the gut microbiome profile, which interacts with immune and inflammatory pathways, both at the systemic and intratumoral levels. As the influence of the gut microbiota on the response to ICI has been repeatedly reported, a specific gut microbiome profile in obese cancer patients may be involved in their better response to ICI. This review summarizes recent data on the interactions between obesity, gut microbiota, and ICIs. In addition, we highlight possible pathophysiological mechanisms supporting the hypothesis that gut microbiota could be one of the links between obesity and poor response to ICIs.
Collapse
Affiliation(s)
- Matthieu Delaye
- Curie Institute, Department of medical oncology, Versailles Saint-Quentin University, Saint-Cloud, France; GERCOR, 75011 Paris, France
| | - Adrien Rousseau
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Léah Mailly-Giacchetti
- Department of Medical Oncology, Saint-Louis Hospital, AP-HP.Nord - Université de Paris, Paris, France
| | - Sandra Assoun
- Department of Thoracic Oncology & CIC 1425/CLIP2 Paris-Nord, Bichat-Claude Bernard Hospital, APHP, Paris, France
| | - Harry Sokol
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France; Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Paris, France; INRAE, AgroParisTech, Micalis Institut, 78350, Jouy-en-Josas, France
| | - Cindy Neuzillet
- Curie Institute, Department of medical oncology, Versailles Saint-Quentin University, Saint-Cloud, France; GERCOR, 75011 Paris, France.
| |
Collapse
|
35
|
Zhao LY, Mei JX, Yu G, Lei L, Zhang WH, Liu K, Chen XL, Kołat D, Yang K, Hu JK. Role of the gut microbiota in anticancer therapy: from molecular mechanisms to clinical applications. Signal Transduct Target Ther 2023; 8:201. [PMID: 37179402 PMCID: PMC10183032 DOI: 10.1038/s41392-023-01406-7] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 02/21/2023] [Accepted: 03/12/2023] [Indexed: 05/15/2023] Open
Abstract
In the past period, due to the rapid development of next-generation sequencing technology, accumulating evidence has clarified the complex role of the human microbiota in the development of cancer and the therapeutic response. More importantly, available evidence seems to indicate that modulating the composition of the gut microbiota to improve the efficacy of anti-cancer drugs may be feasible. However, intricate complexities exist, and a deep and comprehensive understanding of how the human microbiota interacts with cancer is critical to realize its full potential in cancer treatment. The purpose of this review is to summarize the initial clues on molecular mechanisms regarding the mutual effects between the gut microbiota and cancer development, and to highlight the relationship between gut microbes and the efficacy of immunotherapy, chemotherapy, radiation therapy and cancer surgery, which may provide insights into the formulation of individualized therapeutic strategies for cancer management. In addition, the current and emerging microbial interventions for cancer therapy as well as their clinical applications are summarized. Although many challenges remain for now, the great importance and full potential of the gut microbiota cannot be overstated for the development of individualized anti-cancer strategies, and it is necessary to explore a holistic approach that incorporates microbial modulation therapy in cancer.
Collapse
Affiliation(s)
- Lin-Yong Zhao
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jia-Xin Mei
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Gang Yu
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Lei
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University; Frontier Innovation Center for Dental Medicine Plus, Sichuan University, Chengdu, China
| | - Wei-Han Zhang
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Liu
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiao-Long Chen
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Damian Kołat
- Department of Experimental Surgery, Medical University of Lodz, Lodz, Poland
| | - Kun Yang
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Jian-Kun Hu
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
36
|
Gao G, Shen S, Zhang T, Zhang J, Huang S, Sun Z, Zhang H. Lacticaseibacillus rhamnosus Probio-M9 enhanced the antitumor response to anti-PD-1 therapy by modulating intestinal metabolites. EBioMedicine 2023; 91:104533. [PMID: 37027929 PMCID: PMC10085781 DOI: 10.1016/j.ebiom.2023.104533] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 02/23/2023] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Probiotics have been increasingly proposed for enhancing immune checkpoint blockade (ICB) treatments against cancer. However, its causal relationship with immunotherapeutic efficacy remains unclear, which promoted us to explore if and how probiotic Lacticaseibacillus rhamnosus Probio-M9 manipulates gut microbiome for expected outcomes. METHODS We evaluated the effects of Probio-M9 on the anti-PD-1 treatment against colorectal cancer in mice via a multi-omics approach. We defined the mechanisms of Probio-M9-mediated antitumor immunity by comprehensive analyses of metagenome and metabolites of commensal gut microbes as well as the immunologic factors and serum metabolome of the host. FINDINGS The results indicated that Probio-M9 intervention strengthened the anti-PD-1-based tumor inhibition. Both prophylactic and therapeutic administration of Probio-M9 showed conspicuous performance in controlling tumor growth with ICB treatment. The supplement of Probio-M9 modulated enhanced immunotherapy response through promoting beneficial microbes (e.g., Lactobacillus and Bifidobacterium animalis), producing beneficial metabolites including butyric acids in the gut, and accumulating blood-derived α-ketoglutaric acid, N-acetyl-l-glutamic acid and pyridoxine in particular, which promoted the infiltration and activation of cytotoxic T lymphocytes (CTLs) and suppressing the function of regulatory T cells (Tregs) in the tumor microenvironment (TME). Subsequently, we found that enhanced immunotherapeutic response was transmissible by transplanting either post-probiotic-treatment gut microbes or intestinal metabolites to new tumor-bearing mice. INTERPRETATION This study offered valuable insight into the causal role of Probio-M9 in correcting the defects in gut microbiota that compromised anti-PD-1 therapeutic efficacy, which can be used as an alternative synergetic agent with ICB for clinical cancer treatment. FUNDING This research was supported by Research Fund for the National Key R&D Program of China (2022YFD2100702), Inner Mongolia Science and Technology Major Projects (2021ZD0014), and China Agriculture Research System of MOF and MARA.
Collapse
Affiliation(s)
- Guangqi Gao
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Siyuan Shen
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Tao Zhang
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Jiachao Zhang
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Shi Huang
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Zhihong Sun
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Heping Zhang
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China.
| |
Collapse
|
37
|
Pinto C, Aluai-Cunha C, Santos A. The human and animals' malignant melanoma: comparative tumor models and the role of microbiome in dogs and humans. Melanoma Res 2023; 33:87-103. [PMID: 36662668 DOI: 10.1097/cmr.0000000000000880] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Currently, the most progressively occurring incident cancer is melanoma. The mouse is the most popular model in human melanoma research given its various benefits as a laboratory animal. Nevertheless, unlike humans, mice do not develop melanoma spontaneously, so they need to be genetically manipulated. In opposition, there are several reports of other animals, ranging from wild to domesticated animals, that spontaneously develop melanoma and that have cancer pathways that are similar to those of humans. The influence of the gut microbiome on health and disease is being the aim of many recent studies. It has been proven that the microbiome is a determinant of the host's immune status and disease prevention. In human medicine, there is increasing evidence that changes in the microbiome influences malignant melanoma progression and response to therapy. There are several similarities between some animals and human melanoma, especially between canine and human oral malignant melanoma as well as between the gut microbiome of both species. However, microbiome studies are scarce in veterinary medicine, especially in the oncology field. Future studies need to address the relevance of gut and tissue microbiome for canine malignant melanoma development, which results will certainly benefit both species in the context of translational medicine.
Collapse
Affiliation(s)
- Catarina Pinto
- Department of Veterinary Clinics, Institute of Biomedical Sciences Abel Salazar of the University of Porto (ICBAS-UP)
| | - Catarina Aluai-Cunha
- Department of Veterinary Clinics, Institute of Biomedical Sciences Abel Salazar of the University of Porto (ICBAS-UP)
| | - Andreia Santos
- Department of Veterinary Clinics, Institute of Biomedical Sciences Abel Salazar of the University of Porto (ICBAS-UP)
- Animal Science and Study Centre (CECA), Food and Agragrian Sciences and Technologies Institute (ICETA), Apartado, Porto, Portugal
| |
Collapse
|
38
|
Paz Del Socorro T, Tonneau M, Pasquier D, Chamaillard M. Short- and Long-term Repercussions of Vancomycin on Immune Surveillance and the Efficacy of Antitumor Treatments. Cancer J 2023; 29:98-101. [PMID: 36957980 DOI: 10.1097/ppo.0000000000000652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
ABSTRACT Although antibiotic is a major contributor to shifts in the intestinal flora that may persist for up to several months after cessation, it is now increasingly recognized that its prescription may differentially influence clinical outcome of different anticancer treatments. Intense clinical and basic research efforts aim then at gaining sufficient insights about how the cooperative action between the intestinal ecosystem and immune surveillance modulates the efficacy of anticancer treatments. In this review, we summarize multiple levels of knowledge between vancomycin exposure, the gut microbiota, and a meaningful therapeutic response. Furthermore, we discuss the mode of action of antibiotic therapy that is prescribed for prophylaxis of bacteremia and neutropenia and outline the opportunity for judiciously improving the efficacy of anticancer drugs.
Collapse
Affiliation(s)
| | - Marion Tonneau
- Academic Department of Radiation Oncology, Centre Oscar Lambret
| | | | | |
Collapse
|
39
|
Rühle A, Zou J, Glaser M, Halle L, Gkika E, Schäfer H, Knopf A, Becker C, Grosu AL, Popp I, Nicolay NH. The influence of antibiotic administration on the outcomes of head-and-neck squamous cell carcinoma patients undergoing definitive (chemo)radiation. Eur Arch Otorhinolaryngol 2023; 280:2605-2616. [PMID: 36764957 PMCID: PMC10066162 DOI: 10.1007/s00405-023-07868-3] [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: 12/03/2022] [Accepted: 01/31/2023] [Indexed: 02/12/2023]
Abstract
PURPOSE Effects of antibiotic administration on patients' microbiome may negatively influence cancer outcomes, and adverse prognoses after antibiotic application have been demonstrated for cancer patients receiving immune checkpoint inhibitors. While the microbiome may play an important role also in head-and-neck squamous cell carcinoma (HNSCC), the prognostic value of antibiotic treatment here is largely unknown. We therefore analyzed whether antibiotic prescription is associated with impaired oncological outcomes of HNSCC patients undergoing definitive (chemo)radiation. METHODS A cohort of 220 HNSCC patients undergoing definitive (chemo)radiation between 2010 and 2019 was analyzed. The influence of antibiotic administration on locoregional control, progression-free survival (PFS) and overall survival (OS) was determined using Kaplan-Meier and Cox analyses. RESULTS A total of 154 patients were treated with antibiotics within 30 days before (chemo)radiation (pretherapeutic) or during (chemo)radiation (peritherapeutic). While antibiotic prescription was not associated with age, ECOG, tumor localization or radiotherapy characteristics, patients treated with antibiotics had significantly higher tumor stages. Peritherapeutic antibiotic administration diminished PFS (HR = 1.397, p < 0.05, log-rank test) and OS (HR = 1.407, p < 0.05), whereas pretherapeutic administration did not. Antibiotic application was an independent prognosticator for OS (HR = 1.703, p < 0.05) and PFS (HR = 1.550, p < 0.05) in the multivariate Cox analysis within the subgroup of patients aged < 75 years. CONCLUSION Peritherapeutic antibiotic usage was associated with impaired oncological outcomes in HNSCC patients undergoing (chemo)radiation. Further studies including microbiome analyses are required to elucidate underlying mechanisms.
Collapse
Affiliation(s)
- Alexander Rühle
- Department of Radiation Oncology, University of Freiburg-Medical Center, Robert-Koch-Str. 3, 79106, Freiburg, Germany.
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Jiadai Zou
- Department of Radiation Oncology, University of Freiburg-Medical Center, Robert-Koch-Str. 3, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Margaretha Glaser
- Department of Radiation Oncology, University of Freiburg-Medical Center, Robert-Koch-Str. 3, 79106, Freiburg, Germany
| | - Lennard Halle
- Department of Radiation Oncology, University of Freiburg-Medical Center, Robert-Koch-Str. 3, 79106, Freiburg, Germany
| | - Eleni Gkika
- Department of Radiation Oncology, University of Freiburg-Medical Center, Robert-Koch-Str. 3, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Henning Schäfer
- Department of Radiation Oncology, University of Freiburg-Medical Center, Robert-Koch-Str. 3, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Knopf
- Department of Otorhinolaryngology, University of Freiburg-Medical Center, Freiburg, Germany
| | - Christoph Becker
- Department of Otorhinolaryngology, University of Freiburg-Medical Center, Freiburg, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, University of Freiburg-Medical Center, Robert-Koch-Str. 3, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ilinca Popp
- Department of Radiation Oncology, University of Freiburg-Medical Center, Robert-Koch-Str. 3, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, University of Freiburg-Medical Center, Robert-Koch-Str. 3, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
40
|
Williams KC, Gault A, Anderson AE, Stewart CJ, Lamb CA, Speight RA, Rajan N, Plummer R, Pratt AG. Immune-related adverse events in checkpoint blockade: Observations from human tissue and therapeutic considerations. Front Immunol 2023; 14:1122430. [PMID: 36776862 PMCID: PMC9909476 DOI: 10.3389/fimmu.2023.1122430] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/16/2023] [Indexed: 01/27/2023] Open
Abstract
Checkpoint inhibitors (CPIs) are monoclonal antibodies which, by disrupting interactions of immune checkpoint molecules with their ligands, block regulatory immune signals otherwise exploited by cancers. Despite revolutionary clinical benefits, CPI use is associated with an array of immune-related adverse events (irAEs) that mirror spontaneous autoreactivity. Severe irAEs necessitate pausing or stopping of CPI therapy and use of corticosteroids and/or other immunomodulatory interventions. Despite increasingly widespread CPI use, irAE pathobiology remains poorly understood; its elucidation may point to targeted mitigation strategies and uncover predictive biomarkers for irAE onset in patients, whilst casting new light on mechanisms of spontaneous immune-mediated disease. This review focuses on common CPI-induced irAEs of the gut, skin and synovial joints, and how these compare to immune-mediated diseases such as ulcerative colitis, vitiligo and inflammatory arthritis. We review current understanding of the immunological changes reported following CPI therapy at the level of peripheral blood and tissue. Many studies highlight dysregulation of cytokines in irAE-affected tissue, particularly IFNγ and TNF. IrAE-affected tissues are also predominantly infiltrated by T-cells, with low B-cell infiltration. Whilst there is variability between studies, patients treated with anti-programmed cell death-1 (PD-1)/PDL-1 therapies seem to exhibit CD8+ T-cell dominance, with CD4+ T-cells dominating in those treated with anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) monotherapy. Interestingly, CD8+CXCR3+ T-cells have been reported to be elevated in gastrointestinal, dermatological and musculoskeletal -irAE affected tissues. These findings may highlight potential opportunities for therapeutic development or re-deployment of existing therapies to prevent and/or improve the outcome of irAEs.
Collapse
Affiliation(s)
- Kristian C. Williams
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Abigail Gault
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Northern Centre for Cancer Care, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Amy E. Anderson
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christopher J. Stewart
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christopher A. Lamb
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Department of Gastroenterology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - R. Ally Speight
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Department of Gastroenterology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Neil Rajan
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Department of Dermatology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Ruth Plummer
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Northern Centre for Cancer Care, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Arthur G. Pratt
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Directorate of Musculoskeletal Services, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
41
|
Vihinen H, Jokinen A, Laajala TD, Wahid N, Peltola L, Kettunen T, Rönkä A, Tiainen L, Skyttä T, Kohtamäki L, Tulokas S, Karhapää H, Hernberg M, Silvoniemi M, Mattila KE. Antibiotic Treatment is an Independent Poor Risk Factor in NSCLC But Not in Melanoma Patients Who had Received Anti-PD-1/L1 Monotherapy. Clin Lung Cancer 2023; 24:295-304. [PMID: 36774235 DOI: 10.1016/j.cllc.2023.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/26/2023]
Abstract
BACKGROUND Antibiotic treatment may reduce the efficacy of cancer immunotherapy by disrupting gut microbiome. We aimed to study the association of antibiotics and survival outcomes in advanced cutaneous melanoma and non-small-cell lung cancer (NSCLC) patients who had received anti-PD-1/L1 monotherapy. PATIENTS AND METHODS A total of 222 melanoma and 199 NSCLC patients had received anti-PD-1/L1 monotherapy in 5 Finnish hospitals between January 2014 and December 2020. Clinical characteristics, antibiotic and corticosteroid treatment, and survival outcomes were retrospectively collected from hospital and national medical records. RESULTS There were 32% of melanoma and 31% of NSCLC patients who had received antibiotic treatment (ABT) 3 months before to 1 month after the first anti-PD-1/L1 antibody infusion. In survival analyses, early antibiotic treatment was associated with inferior overall survival (OS) (ABT 19.2 [17.6-43.7] vs. no ABT 35.6 [29.3-NA] months, P = .033) but not with inferior progression-free survival (PFS) (ABT 5.8 [3.0-12.6] vs. no ABT 10.2 [7.7-15.3] months, P = .3) in melanoma patients and with inferior OS (ABT 8.6 [6.4-12.3] vs. no ABT 18.5 [15.1-21.6] months, P < .001) and PFS (ABT 2.8 [2.1-4.5] vs. no ABT 5.6 [4.4-8.0] months, P = .0081) in NSCLC patients. In multivariable analyses, ABT was not an independent risk-factor for inferior OS and PFS in melanoma but was associated with inferior OS (hazard ratio [HR] 2.12 [1.37-3.28]) and PFS (HR 1.65 [1.10-2.47]) in NSCLC after adjusted for other risk factors. CONCLUSIONS Early ABT was an independent poor risk factor in NSCLC patients who had received anti-PD-1/L1 monotherapy but not in melanoma patients. The weight of ABT as a poor risk factor might depend on other prognostic factors in different cancers.
Collapse
Affiliation(s)
- Hannes Vihinen
- Department of Oncology and Radiotherapy and Fican West Cancer Centre, University of Turku and Turku University Hospital, POB 52, FIN-20521 Turku, Finland
| | - Artturi Jokinen
- Department of Oncology and Radiotherapy and Fican West Cancer Centre, University of Turku and Turku University Hospital, POB 52, FIN-20521 Turku, Finland
| | - Teemu D Laajala
- Department of Mathematics and Statistics, University of Turku, Turku, Finland
| | - Nesna Wahid
- Department of Oncology and Radiotherapy Vaasa Central Hospital, Vaasa Finland
| | - Lotta Peltola
- Department of Oncology and Radiotherapy Vaasa Central Hospital, Vaasa Finland
| | - Tiia Kettunen
- Center of Oncology, Kuopio University Hospital, Kuopio, Finland
| | - Aino Rönkä
- Center of Oncology, Kuopio University Hospital, Kuopio, Finland
| | - Leena Tiainen
- Department of Oncology, Tays Cancer Centre, Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Tanja Skyttä
- Department of Oncology, Tays Cancer Centre, Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Laura Kohtamäki
- Comprehensive Cancer Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Sanni Tulokas
- Comprehensive Cancer Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Hanna Karhapää
- Comprehensive Cancer Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Micaela Hernberg
- Comprehensive Cancer Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Maria Silvoniemi
- Department of Respiratory Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Kalle E Mattila
- Department of Oncology and Radiotherapy and Fican West Cancer Centre, University of Turku and Turku University Hospital, POB 52, FIN-20521 Turku, Finland; InFLAMES Research Flagship Center, University of Turku.
| |
Collapse
|
42
|
Wang Y, Jenq RR, Wargo JA, Watowich SS. Microbiome influencers of checkpoint blockade-associated toxicity. J Exp Med 2023; 220:213796. [PMID: 36622383 PMCID: PMC9836236 DOI: 10.1084/jem.20220948] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/29/2022] [Accepted: 12/09/2022] [Indexed: 01/10/2023] Open
Abstract
Immunotherapy has greatly improved cancer outcomes, yet variability in response and off-target tissue damage can occur with these treatments, including immune checkpoint inhibitors (ICIs). Multiple lines of evidence indicate the host microbiome influences ICI response and risk of immune-related adverse events (irAEs). As the microbiome is modifiable, these advances indicate the potential to manipulate microbiome components to increase ICI success. We discuss microbiome features associated with ICI response, with focus on bacterial taxa and potential immune mechanisms involved in irAEs, and the overall goal of driving novel approaches to manipulate the microbiome to improve ICI efficacy while avoiding irAE risk.
Collapse
Affiliation(s)
- Yinghong Wang
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert R. Jenq
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Platform for Innovative Microbiome and Translational Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer A. Wargo
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Platform for Innovative Microbiome and Translational Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephanie S. Watowich
- Platform for Innovative Microbiome and Translational Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
43
|
Pirini F, Cortesi M, Tumedei MM, Zanoni M, Ravaioli S, Bravaccini S. Tumor resident microbiota and response to therapies: An insight on tissue bacterial microbiota. Front Cell Dev Biol 2023; 10:1048360. [PMID: 36684442 PMCID: PMC9845623 DOI: 10.3389/fcell.2022.1048360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/15/2022] [Indexed: 01/06/2023] Open
Abstract
The role of the intestinal microbiota in the promotion, progression, and response to therapies is gaining importance, but recent studies confirm the presence of microbiota also in the tumor, thus becoming a component of the tumor microenvironment. There is not much knowledge on the characteristics and mechanisms of action of the tumor resident microbiota, but there are already indications of its involvement in conditioning the response to therapies. In this review, we discuss recent publications on the interaction between microbiota and anticancer treatments, mechanisms of resistance and possible strategies for manipulating the microbiota that could improve treatments in a personalized medicine perspective.
Collapse
|
44
|
Wang G, He X, Wang Q. Intratumoral bacteria are an important "accomplice" in tumor development and metastasis. Biochim Biophys Acta Rev Cancer 2023; 1878:188846. [PMID: 36496095 DOI: 10.1016/j.bbcan.2022.188846] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/09/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022]
Abstract
As emerging tumor components, intratumoral bacteria have been found in many solid tumors. Several studies have demonstrated that different cancer subtypes have distinct microbial compositions, and mechanistic studies have shown that intratumoral bacteria may promote cancer initiation and progression through DNA damage, epigenetic modification, inflammatory responses, modulation of host immunity and activation of oncogenes or oncogenic pathways. Moreover, intratumoral bacteria have been shown to modulate tumor metastasis and chemotherapy response. A better understanding of the tumor microenvironment and its associated microbiota will facilitate the design of new metabolically engineered species, opening up a new era of intratumoral bacteria-based cancer therapy. However, many questions remain to be resolved, such as where intratumoral bacteria originate and whether there is a direct causal relationship between intratumoral bacteria and tumor susceptibility. In addition, suitable preclinical models and more advanced detection techniques are crucial for studying the biological functions of intratumoral bacteria. In this review, we summarize the complicated role of intratumoral bacteria in the regulation of cancer development and metastasis and discuss their carcinogenic mechanisms and potential therapeutic aspects.
Collapse
Affiliation(s)
- Gang Wang
- Department of General Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China; Department of General Surgery, The 74th Group Army Hospital, Guangzhou 510318, China
| | - Xianli He
- Department of General Surgery, Tangdu Hospital, Air Force Military Medical University, Xi'an 710032, Shaanxi, China.
| | - Qian Wang
- Department of General Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
45
|
Shi Z, Li H, Song W, Zhou Z, Li Z, Zhang M. Emerging roles of the gut microbiota in cancer immunotherapy. Front Immunol 2023; 14:1139821. [PMID: 36911704 PMCID: PMC9992551 DOI: 10.3389/fimmu.2023.1139821] [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: 01/07/2023] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
Gut microbiota represents a hidden treasure vault encompassing trillions of microorganisms that inhabit the intestinal epithelial barrier of the host. In the past decade, numerous in-vitro, animal and clinical studies have revealed the profound roles of gut microbiota in maintaining the homeostasis of various physiological functions, especially immune modulation, and remarkable differences in the configuration of microbial communities between cancers and healthy individuals. In addition, although considerable efforts have been devoted to cancer treatments, there remain many patients succumb to their disease with the incremental cancer burden worldwide. Nevertheless, compared with the stability of human genome, the plasticity of gut microbiota renders it a promising opportunity for individualized treatment. Meanwhile, burgeoning findings indicate that gut microbiota is involved in close interactions with the outcomes of diverse cancer immunotherapy protocols, including immune checkpoint blockade therapy, allogeneic hematopoietic stem cell transplantation, and chimeric antigen receptor T cell therapy. Here, we reviewed the evidence for the capacity of gut microflora to modulate cancer immunotherapies, and highlighted the opportunities of microbiota-based prognostic prediction, as well as microbiotherapy by targeting the microflora to potentiate anticancer efficacy while attenuating toxicity, which will be pivotal to the development of personalized cancer treatment strategies.
Collapse
Affiliation(s)
- Zhuangzhuang Shi
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, China.,Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongwen Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, China
| | - Wenting Song
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, China.,Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhiyuan Zhou
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, China
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
46
|
Crespin A, Le Bescop C, de Gunzburg J, Vitry F, Zalcman G, Cervesi J, Bandinelli PA. A systematic review and meta-analysis evaluating the impact of antibiotic use on the clinical outcomes of cancer patients treated with immune checkpoint inhibitors. Front Oncol 2023; 13:1075593. [PMID: 36937417 PMCID: PMC10019357 DOI: 10.3389/fonc.2023.1075593] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have considerably improved patient outcomes in various cancer types, but their efficacy remains poorly predictable among patients. The intestinal microbiome, whose balance and composition can be significantly altered by antibiotic use, has recently emerged as a factor that may modulate ICI efficacy. The objective of this systematic review and meta-analysis is to investigate the impact of antibiotics on the clinical outcomes of cancer patients treated with ICIs. Methods PubMed and major oncology conference proceedings were systematically searched to identify all studies reporting associations between antibiotic use and at least one of the following endpoints: Overall Survival (OS), Progression-Free Survival (PFS), Objective Response Rate (ORR) and Progressive Disease (PD) Rate. Pooled Hazard Ratios (HRs) for OS and PFS, and pooled Odds Ratios (ORs) for ORR and PD were calculated. Subgroup analyses on survival outcomes were also performed to investigate the potential differential effect of antibiotics according to cancer types and antibiotic exposure time windows. Results 107 articles reporting data for 123 independent cohorts were included, representing a total of 41,663 patients among whom 11,785 (28%) received antibiotics around ICI initiation. The pooled HRs for OS and PFS were respectively of 1.61 [95% Confidence Interval (CI) 1.48-1.76] and 1.45 [95% CI 1.32-1.60], confirming that antibiotic use was significantly associated with shorter survival. This negative association was observed consistently across all cancer types for OS and depending on the cancer type for PFS. The loss of survival was particularly strong when antibiotics were received shortly before or after ICI initiation. The pooled ORs for ORR and PD were respectively of 0.59 [95% CI 0.47-0.76] and 1.86 [95% CI 1.41-2.46], suggesting that antibiotic use was significantly associated with worse treatment-related outcomes. Conclusion As it is not ethically feasible to conduct interventional, randomized, controlled trials in which antibiotics would be administered to cancer patients treated with ICIs to demonstrate their deleterious impact versus control, prospective observational studies and interventional trials involving microbiome modifiers are crucially needed to uncover the role of microbiome and improve patient outcomes. Such studies will reduce the existing publication bias by allowing analyses on more homogeneous populations, especially in terms of treatments received, which is not possible at this stage given the current state of the field. In the meantime, antibiotic prescription should be cautiously considered in cancer patients receiving ICIs. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42019145675.
Collapse
Affiliation(s)
- Athéna Crespin
- Da Volterra, Paris, France
- *Correspondence: Athéna Crespin,
| | | | | | | | - Gérard Zalcman
- Department of Thoracic Oncology and CIC1425, Institut du Cancer AP-HP, Nord, Hôpital Bichat-Claude Bernard, AP-HP, Université de Paris, Paris, France
- U830 Institut National de la Santé et de la Recherche Médicale (INSERM) “Cancer, Heterogeneity, Instability and Plasticity” Curie Institute, Paris, France
| | | | | |
Collapse
|
47
|
Borgers JSW, Burgers FH, Terveer EM, van Leerdam ME, Korse CM, Kessels R, Flohil CC, Blank CU, Schumacher TN, van Dijk M, Henderickx JGE, Keller JJ, Verspaget HW, Kuijper EJ, Haanen JBAG. Conversion of unresponsiveness to immune checkpoint inhibition by fecal microbiota transplantation in patients with metastatic melanoma: study protocol for a randomized phase Ib/IIa trial. BMC Cancer 2022; 22:1366. [PMID: 36585700 PMCID: PMC9801532 DOI: 10.1186/s12885-022-10457-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/16/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The gut microbiome plays an important role in immune modulation. Specifically, presence or absence of certain gut bacterial taxa has been associated with better antitumor immune responses. Furthermore, in trials using fecal microbiota transplantation (FMT) to treat melanoma patients unresponsive to immune checkpoint inhibitors (ICI), complete responses (CR), partial responses (PR), and durable stable disease (SD) have been observed. However, the underlying mechanism determining which patients will or will not respond and what the optimal FMT composition is, has not been fully elucidated, and a discrepancy in microbial taxa associated with clinical response has been observed between studies. Furthermore, it is unknown whether a change in the microbiome itself, irrespective of its origin, or FMT from ICI responding donors, is required for reversion of ICI-unresponsiveness. To address this, we will transfer microbiota of either ICI responder or nonresponder metastatic melanoma patients via FMT. METHODS In this randomized, double-blinded phase Ib/IIa trial, 24 anti-PD1-refractory patients with advanced stage cutaneous melanoma will receive an FMT from either an ICI responding or nonresponding donor, while continuing anti-PD-1 treatment. Donors will be selected from patients with metastatic melanoma treated with anti-PD-1 therapy. Two patients with a good response (≥ 30% decrease according to RECIST 1.1 within the past 24 months) and two patients with progression (≥ 20% increase according to RECIST 1.1 within the past 3 months) will be selected as ICI responding or nonresponding donors, respectively. The primary endpoint is clinical benefit (SD, PR or CR) at 12 weeks, confirmed on a CT scan at 16 weeks. The secondary endpoint is safety, defined as the occurrence of grade ≥ 3 toxicity. Exploratory endpoints are progression-free survival and changes in the gut microbiome, metabolome, and immune cells. DISCUSSION Transplanting fecal microbiota to restore the patients' perturbed microbiome has proven successful in several indications. However, less is known about the potential role of FMT to improve antitumor immune response. In this trial, we aim to investigate whether administration of FMT can reverse resistance to anti-PD-1 treatment in patients with advanced stage melanoma, and whether the ICI-responsiveness of the feces donor is associated with its effectiveness. TRIAL REGISTRATION ClinicalTrials.gov: NCT05251389 (registered 22-Feb-2022). Protocol V4.0 (08-02-2022).
Collapse
Affiliation(s)
- J. S. W. Borgers
- grid.430814.a0000 0001 0674 1393Department of Medical Oncology, Antoni Van Leeuwenhoek, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - F. H. Burgers
- grid.430814.a0000 0001 0674 1393Department of Medical Oncology, Antoni Van Leeuwenhoek, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - E. M. Terveer
- grid.10419.3d0000000089452978Netherlands Donor Feces Bank, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands ,grid.10419.3d0000000089452978Center for Microbiome Analyses and Therapeutics at Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - M. E. van Leerdam
- grid.430814.a0000 0001 0674 1393Department of Gastrointestinal Oncology, Antoni Van Leeuwenhoek, The Netherlands Cancer Institute, Amsterdam, The Netherlands ,grid.10419.3d0000000089452978Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - C. M. Korse
- grid.430814.a0000 0001 0674 1393Department of Laboratory Medicine, Antoni Van Leeuwenhoek, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - R. Kessels
- grid.430814.a0000 0001 0674 1393Department of Biometrics, Antoni Van Leeuwenhoek, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - C. C. Flohil
- grid.430814.a0000 0001 0674 1393Department of Pathology, Antoni Van Leeuwenhoek, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - C. U. Blank
- grid.430814.a0000 0001 0674 1393Department of Medical Oncology, Antoni Van Leeuwenhoek, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - T. N. Schumacher
- grid.430814.a0000 0001 0674 1393Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands ,grid.10419.3d0000000089452978Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - M. van Dijk
- grid.430814.a0000 0001 0674 1393Clinical Trial Service Unit, Antoni Van Leeuwenhoek, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J. G. E. Henderickx
- grid.10419.3d0000000089452978Center for Microbiome Analyses and Therapeutics at Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - J. J. Keller
- grid.10419.3d0000000089452978Netherlands Donor Feces Bank, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands ,grid.10419.3d0000000089452978Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands ,grid.414842.f0000 0004 0395 6796Department of Gastroenterology, Haaglanden Medical Center, Den Haag, The Netherlands
| | - H. W. Verspaget
- grid.10419.3d0000000089452978Netherlands Donor Feces Bank, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands ,grid.10419.3d0000000089452978Department of Biobanking, Leiden University Medical Center, Leiden, The Netherlands
| | - E. J. Kuijper
- grid.10419.3d0000000089452978Netherlands Donor Feces Bank, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands ,grid.10419.3d0000000089452978Center for Microbiome Analyses and Therapeutics at Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - J. B. A. G. Haanen
- grid.430814.a0000 0001 0674 1393Department of Medical Oncology, Antoni Van Leeuwenhoek, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
48
|
Yu J, Yin Y, Yu Y, Cheng M, Zhang S, Jiang S, Dong M. Effect of concomitant antibiotics use on patient outcomes and adverse effects in patients treated with ICIs. Immunopharmacol Immunotoxicol 2022; 45:386-394. [PMID: 36382735 DOI: 10.1080/08923973.2022.2145966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Jiuhang Yu
- College of Pharmacy, Jiamusi University, Jiamusi, China
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yichuang Yin
- College of Pharmacy, Jiamusi University, Jiamusi, China
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yang Yu
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Mengfei Cheng
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shuo Zhang
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shuai Jiang
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Mei Dong
- College of Pharmacy, Jiamusi University, Jiamusi, China
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
49
|
Zhou G, Zhang N, Meng K, Pan F. Interaction between gut microbiota and immune checkpoint inhibitor-related colitis. Front Immunol 2022; 13:1001623. [PMID: 36389768 PMCID: PMC9648670 DOI: 10.3389/fimmu.2022.1001623] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/10/2022] [Indexed: 09/23/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have become a promising therapeutic strategy for malignant tumors, improving patient prognosis, along with a spectrum of immune-related adverse events (irAEs), including gastrointestinal toxicity, ICI-related colitis (IRC), and diarrhea. The gut microbiota has been suggested as an important regulator in the pathogenesis of IRC, and microbiota modulations like probiotics and fecal microbiota transplantation have been explored to treat the disease. This review discusses the interaction between the gut microbiota and IRC, focusing on the potential pathogenic mechanisms and promising interventions.
Collapse
Affiliation(s)
- Guanzhou Zhou
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Nana Zhang
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Ke Meng
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Fei Pan
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
50
|
Han X, Zang D, Liu D, Chen J. The multifaceted roles of common gut microbiota in immune checkpoint inhibitor-mediated colitis: From mechanism to clinical application. Front Immunol 2022; 13:988849. [PMID: 36189293 PMCID: PMC9515466 DOI: 10.3389/fimmu.2022.988849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
With the arrival of the era of tumor immunotherapy, Immune Checkpoint Inhibitors have benefited countless tumor patients. However, the emergence of Immune-Related Adverse Events, especially Immune Checkpoint Inhibitor-Mediated Colitis (IMC), has become an important obstacle to immunotherapy. Therefore, it is very important to clarify the mechanism and influencing factors of IMC. The effect of gut microbiota on IMC is gradually becoming a research hotspot. Gut microbiota from different phyla can affect IMC by regulating innate and acquired immunity of tumor patients in various ways. In this review, we make a systematic and comprehensive introduction of the effect of gut microbiota on IMC. Through understanding the specific effects of gut microbiota on IMC, and then exploring the possibility of reducing IMC by regulating gut microbiota.
Collapse
|