51
|
Eng L, Sutradhar R, Niu Y, Liu N, Liu Y, Kaliwal Y, Powis ML, Liu G, Peppercorn JM, Bedard PL, Krzyzanowska MK. Impact of Antibiotic Exposure Before Immune Checkpoint Inhibitor Treatment on Overall Survival in Older Adults With Cancer: A Population-Based Study. J Clin Oncol 2023:JCO2200074. [PMID: 36827626 DOI: 10.1200/jco.22.00074] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023] Open
Abstract
PURPOSE Antibiotic exposure before immune checkpoint inhibitor (ICI) treatment can negatively affect outcomes through alteration in the gut microbiome, but large-scale evaluations are lacking. We performed a population-level retrospective cohort study to evaluate the impact of antibiotic exposure before starting ICI on overall survival (OS). PATIENT AND METHODS Patients with cancer, age 65 years or older, who initiated treatment with ICIs between June 2012 and October 2018 in Ontario, Canada, were identified using systemic therapy administration data. The cohort was deterministically linked to other health care databases to obtain covariates and antibiotic prescription claim data at both 1 year and 60 days before ICI therapy. Multivariable Cox models evaluated the association between exposure and OS. RESULTS Among the 2,737 patients with cancer who received ICIs, 59% and 19% of patients received antibiotics 1 year and 60 days before ICI therapy, respectively. Median OS was 306 days. Any antibiotic exposure within 1 year before ICI was associated with worse OS (adjusted hazard ratio [aHR], 1.12; 95% CI, 1.12 to 1.23; P = .03). In antibiotic class analysis, exposure to fluoroquinolones within 1 year (aHR, 1.26; 95% CI, 1.13 to 1.40; P < .001) or 60 days before ICI (aHR, 1.20; 95% CI, 0.99 to 1.45; P = .06) was associated with worse OS, with a dose effect seen on the basis of total weeks of exposure over 1 year (aHR, 1.07 per week; 95% CI, 1.03 to 1.11; P < .001) and 60 days (aHR, 1.12 per week; 95% CI, 1.03 to 1.23; P = .01). CONCLUSION In this population-level study, exposure to antibiotics and specifically fluoroquinolones before ICI therapy was observed to be associated with worse OS among older adults with cancer. Interventions aimed at altering the gut microbiome to boost immunogenicity may help improve outcomes for patients receiving ICIs with prior antibiotic exposure.
Collapse
Affiliation(s)
- Lawson Eng
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Divison of Medical Oncology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Rinku Sutradhar
- Cancer Research Program, Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada.,Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Yue Niu
- Cancer Research Program, Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| | - Ning Liu
- Cancer Research Program, Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| | - Ying Liu
- Cancer Research Program, Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| | - Yosuf Kaliwal
- Cancer Research Program, Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| | - Melanie L Powis
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Geoffrey Liu
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Divison of Medical Oncology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey M Peppercorn
- Division of Hematology/Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Philippe L Bedard
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Divison of Medical Oncology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Monika K Krzyzanowska
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Divison of Medical Oncology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,Cancer Research Program, Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| |
Collapse
|
52
|
Yuan H, Gui R, Wang Z, Fang F, Zhao H. Gut microbiota: A novel and potential target for radioimmunotherapy in colorectal cancer. Front Immunol 2023; 14:1128774. [PMID: 36798129 PMCID: PMC9927011 DOI: 10.3389/fimmu.2023.1128774] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/17/2023] [Indexed: 02/04/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers, with a high mortality rate, and is a major burden on human health worldwide. Gut microbiota regulate human immunity and metabolism through producing numerous metabolites, which act as signaling molecules and substrates for metabolic reactions in various biological processes. The importance of host-gut microbiota interactions in immunometabolic mechanisms in CRC is increasingly recognized, and interest in modulating the microbiota to improve patient's response to therapy has been raising. However, the specific mechanisms by which gut microbiota interact with immunotherapy and radiotherapy remain incongruent. Here we review recent advances and discuss the feasibility of gut microbiota as a regulatory target to enhance the immunogenicity of CRC, improve the radiosensitivity of colorectal tumor cells and ameliorate complications such as radiotoxicity. Currently, great breakthroughs in the treatment of non-small cell lung cancer and others have been achieved by radioimmunotherapy, but radioimmunotherapy alone has not been effective in CRC patients. By summarizing the recent preclinical and clinical evidence and considering regulatory roles played by microflora in the gut, such as anti-tumor immunity, we discuss the potential of targeting gut microbiota to enhance the efficacy of radioimmunotherapy in CRC and expect this review can provide references and fresh ideas for the clinical application of this novel strategy.
Collapse
Affiliation(s)
- Hanghang Yuan
- Department of Nuclear Medicine, The First Hospital of Jilin University, Changchun, China,National Health Commission (NHC) Key laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Ruirui Gui
- Department of Nuclear Medicine, The First Hospital of Jilin University, Changchun, China,National Health Commission (NHC) Key laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Zhicheng Wang
- National Health Commission (NHC) Key laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Fang Fang
- National Health Commission (NHC) Key laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China,*Correspondence: Fang Fang, ; Hongguang Zhao,
| | - Hongguang Zhao
- Department of Nuclear Medicine, The First Hospital of Jilin University, Changchun, China,*Correspondence: Fang Fang, ; Hongguang Zhao,
| |
Collapse
|
53
|
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: 8.0] [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
|
54
|
Gonugunta AS, Von Itzstein MS, Hsiehchen D, Le T, Rashdan S, Yang H, Selby C, Alvarez C, Gerber DE. Antibiotic Prescriptions in Lung Cancer and Melanoma Populations: Differences With Potential Clinical Implications in the Immunotherapy Era. Clin Lung Cancer 2023; 24:11-17. [PMID: 36253271 DOI: 10.1016/j.cllc.2022.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 09/14/2022] [Accepted: 09/17/2022] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Antibiotic exposure is associated with worse clinical outcomes in patients receiving immune checkpoint inhibitors (ICI). We analyzed antibiotic prescription patterns in lung cancer and melanoma, two malignancies in which ICI are used broadly across stages. METHODS We performed a retrospective cohort study of adults in the U.S. Veterans Affairs (VA) medical system diagnosed with lung cancer or melanoma from 2003 to 2016. We defined antibiotic exposure as receipt of a prescription for a systemic antibacterial agent between 6 months before and 6 months after cancer diagnosis. Demographics, clinical variables, prescriptions, and diagnostic codes were abstracted from the VA Corporate Data Warehouse. Antibiotic exposure was compared using t tests, Chi-square, and multivariate analyses. RESULTS A total of 310,321 patients (280,068 lung cancer, 30,253 melanoma) were included in the analysis. Antibiotic exposure was more common among patients with lung cancer (42% vs. 24% for melanoma; P < .001). Among antibiotic-exposed patients, those with lung cancer were more likely to receive prescriptions for multiple antibiotics (47% vs. 30% for melanoma; P < .001). In multivariate analyses, antibiotic exposure was associated with lung cancer diagnosis (HR 1.50; 95% CI, 1.46-1.55), comorbidity score (HR 1.08; 95% CI, 1.08-1.09), non-white race (HR 1.11; 95% CI, 1.06-1.17), and female gender (HR 1.31; 95% CI, 1.24-1.37). CONCLUSION Among cancer patients, antibiotics are prescribed frequently. Antibiotic exposure is more common in certain cancer types and patient populations. Given the negative effect antibiotic exposure has on immunotherapy outcomes, these observations may have clinical and healthy policy implications.
Collapse
Affiliation(s)
| | - Mitchell S Von Itzstein
- Department of Internal Medicine (Division of Hematology-Oncology), UT Southwestern Medical Center; Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center
| | - David Hsiehchen
- Department of Internal Medicine (Division of Hematology-Oncology), UT Southwestern Medical Center; Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center
| | - Tri Le
- Department of Internal Medicine (Division of Hematology-Oncology), UT Southwestern Medical Center; Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center
| | - Sawsan Rashdan
- Department of Internal Medicine (Division of Hematology-Oncology), UT Southwestern Medical Center; Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center
| | - Hui Yang
- Texas Tech University School of Pharmacy
| | | | - Carlos Alvarez
- Texas Tech University School of Pharmacy; Department of Population and Data Sciences, UT Southwestern Medical Center
| | - David E Gerber
- Department of Internal Medicine (Division of Hematology-Oncology), UT Southwestern Medical Center; Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center; Department of Population and Data Sciences, UT Southwestern Medical Center.
| |
Collapse
|
55
|
Impact of antibiotic use before definitive concurrent chemoradiation in patients with locally advanced non-small cell lung cancer. Strahlenther Onkol 2022:10.1007/s00066-022-02027-9. [DOI: 10.1007/s00066-022-02027-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 11/06/2022] [Indexed: 12/13/2022]
|
56
|
Fernandes MR, Aggarwal P, Costa RGF, Cole AM, Trinchieri G. Targeting the gut microbiota for cancer therapy. Nat Rev Cancer 2022; 22:703-722. [PMID: 36253536 DOI: 10.1038/s41568-022-00513-x] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/07/2022] [Indexed: 02/06/2023]
Abstract
Growing evidence suggests that the gut microbiota modulates the efficacy and toxicity of cancer therapy, most notably immunotherapy and its immune-related adverse effects. The poor response to immunotherapy in patients treated with antibiotics supports this influential role of the microbiota. Until recently, results pertaining to the identification of the microbial species responsible for these effects were incongruent, and relatively few studies analysed the underlying mechanisms. A better understanding of the taxonomy of the species involved and of the mechanisms of action has since been achieved. Defined bacterial species have been shown to promote an improved response to immune-checkpoint inhibitors by producing different products or metabolites. However, a suppressive effect of Gram-negative bacteria may be dominant in some unresponsive patients. Machine learning approaches trained on the microbiota composition of patients can predict the ability of patients to respond to immunotherapy with some accuracy. Thus, interest in modulating the microbiota composition to improve patient responsiveness to therapy has been mounting. Clinical proof-of-concept studies have demonstrated that faecal microbiota transplantation or dietary interventions might be utilized clinically to improve the success rate of immunotherapy in patients with cancer. Here, we review recent advances and discuss emerging strategies for microbiota-based cancer therapies.
Collapse
Affiliation(s)
- Miriam R Fernandes
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Poonam Aggarwal
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Raquel G F Costa
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Alicia M Cole
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Giorgio Trinchieri
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
57
|
Fu A, Yao B, Dong T, Cai S. Emerging roles of intratumor microbiota in cancer metastasis. Trends Cell Biol 2022:S0962-8924(22)00258-6. [DOI: 10.1016/j.tcb.2022.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/22/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022]
|
58
|
Campoa E, Teixeira J, Luz P, Magalhaes J. End-of-Life Antibiotherapy: Factors Associated With Prescription. Cureus 2022; 14:e31634. [DOI: 10.7759/cureus.31634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2022] [Indexed: 11/19/2022] Open
|
59
|
Gut Microbiota Host-Gene Interaction. Int J Mol Sci 2022; 23:ijms232213717. [PMID: 36430197 PMCID: PMC9698405 DOI: 10.3390/ijms232213717] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/28/2022] [Accepted: 11/07/2022] [Indexed: 11/09/2022] Open
Abstract
Studies carried out in the last ten years have shown that the metabolites made up from the gut microbiota are essential for multiple functions, such as the correct development of the immune system of newborns, interception of pathogens, and nutritional enrichment of the diet. Therefore, it is not surprising that alteration of the gut microbiota is the starting point of gastrointestinal infection, obesity, type 2 diabetes, inflammatory bowel disease, colorectal cancer, and lung cancer. Diet changes and antibiotics are the major factors damaging the gut microbiota. Early exposure of the newborns to antibiotics may prevent their correct development of the immune system, exposing them to pathogen infections, allergies, and chronic inflammatory diseases. We already know much on how host genes, microbiota, and the environment interact, owing to experiments in several model animals, especially in mice; advances in molecular technology; microbiota transplantation; and comparative metagenomic analysis. However, much more remains to be known. Longitudinal studies on patients undergoing to therapy, along with the identification of bacteria prevalent in responding patients may provide valuable data for improving therapies.
Collapse
|
60
|
Serpas Higbie V, Rogers J, Hwang H, Qiao W, Xiao L, Dasari A, Mola-Rudd K, Morris VK, Wolff RA, Raghav K, Huey R, Parseghian C, Willis J, Kopetz S, Overman MJ, Johnson B. Antibiotic Exposure Does Not Impact Immune Checkpoint Blockade Response in MSI-H/dMMR Metastatic Colorectal Cancer: A Single-Center Experience. Oncologist 2022; 27:952-957. [PMID: 35946836 PMCID: PMC9632313 DOI: 10.1093/oncolo/oyac162] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/28/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Immune checkpoint blockade (ICB) has improved outcomes for patients with microsatellite instability high (MSI-H)/deficient mismatch repair (dMMR) tumors. However, not all MSI-H/dMMR patients will exhibit the same ICB efficacy. Previous studies suggest that concomitant antibiotic use while receiving ICB may result in poorer outcomes. We aimed to evaluate this association in patients with MSI-H/dMMR metastatic colorectal cancer (mCRC). MATERIALS AND METHODS A single-site, retrospective review of 57 patients with MSI-H/dMMR mCRC that received ICB was completed. Data collected included patient demographics, ICB information, and antibiotic use. Antibiotic exposure was considered from 90 days prior to ICB through 6 weeks after initiation. Primary endpoint was overall response rate (ORR). RESULTS The majority of patients received pembrolizumab (27 [47%]) or nivolumab (17 [30%]) monotherapy as their ICB agent. Of the 57 patients, 19 (33.3%) had antibiotic exposure from 90 days prior to ICB initiation through 6 weeks after initiation with most (13 [68%]) having antibiotic use in the 30 days preceding ICB initiation. Similar ORRs were seen in both groups (P-value > .99). No difference was observed in OS (P-value .29) or PFS (P-value .36) between groups. CONCLUSION Our data show no association of lower response rates or survival in those MSI-H/dMMR patients with mCRC who receive antibiotics around the initiation of ICB. This information needs to be confirmed in a larger prospective cohort.
Collapse
Affiliation(s)
- Victoria Serpas Higbie
- Hematology/Oncology Fellowship, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jane Rogers
- Pharmacy Clinical Services, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hyunsoo Hwang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wei Qiao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lianchun Xiao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Arvind Dasari
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kerri Mola-Rudd
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Van K Morris
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert A Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kanwal Raghav
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ryan Huey
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christine Parseghian
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason Willis
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael J Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Benny Johnson
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
61
|
Shigehiro T, Ueno M, Kijihira M, Takahashi R, Umemura C, Taha EA, Kurosaka C, Asayama M, Murakami H, Satoh A, Nakamura Y, Futami J, Masuda J. Immune State Conversion of the Mesenteric Lymph Node in a Mouse Breast Cancer Model. Int J Mol Sci 2022; 23:ijms231911035. [PMID: 36232335 PMCID: PMC9570492 DOI: 10.3390/ijms231911035] [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: 08/09/2022] [Revised: 09/17/2022] [Accepted: 09/18/2022] [Indexed: 11/16/2022] Open
Abstract
Secondary lymphoid tissues, such as the spleen and lymph nodes (LNs), contribute to breast cancer development and metastasis in both anti- and pro-tumoral directions. Although secondary lymphoid tissues have been extensively studied, very little is known about the immune conversion in mesenteric LNs (mLNs) during breast cancer development. Here, we demonstrate inflammatory immune conversion of mLNs in a metastatic 4T1 breast cancer model. Splenic T cells were significantly decreased and continuously suppressed IFN-γ production during tumor development, while myeloid-derived suppressor cells (MDSCs) were dramatically enriched. However, T cell numbers in the mLN did not decrease, and the MDSCs only moderately increased. T cells in the mLN exhibited conversion from a pro-inflammatory state with high IFN-γ expression to an anti-inflammatory state with high expression of IL-4 and IL-10 in early- to late-stages of breast cancer development. Interestingly, increased migration of CD103+CD11b+ dendritic cells (DCs) into the mLN, along with increased (1→3)-β-D-glucan levels in serum, was observed even in late-stage breast cancer. This suggests that CD103+CD11b+ DCs could prime cancer-reactive T cells. Together, the data indicate that the mLN is an important lymphoid tissue contributing to breast cancer development.
Collapse
Affiliation(s)
- Tsukasa Shigehiro
- Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Correspondence: (T.S.); (J.M.); Tel.: +81-47-121-4060 (T.S.); +81-86-251-8003 (J.M.)
| | - Maho Ueno
- Department of Applied Chemistry and Biotechnology, Faculty of Engineering, Okayama University, Okayama 700-8530, Japan
| | - Mayumi Kijihira
- Department of Applied Chemistry and Biotechnology, Faculty of Engineering, Okayama University, Okayama 700-8530, Japan
| | - Ryotaro Takahashi
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
| | - Chiho Umemura
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Eman A. Taha
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Chisaki Kurosaka
- Department of Applied Chemistry and Biotechnology, Faculty of Engineering, Okayama University, Okayama 700-8530, Japan
| | - Megumi Asayama
- Department of Applied Chemistry and Biotechnology, Faculty of Engineering, Okayama University, Okayama 700-8530, Japan
| | - Hiroshi Murakami
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Ayano Satoh
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Yoshimasa Nakamura
- Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan
| | - Junichiro Futami
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Junko Masuda
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
- Department of Pharmacology, Tokyo Women’s Medical University, Shinjuku, Tokyo 162-8666, Japan
- Correspondence: (T.S.); (J.M.); Tel.: +81-47-121-4060 (T.S.); +81-86-251-8003 (J.M.)
| |
Collapse
|
62
|
Sommerfelt H, Sandvik LF, Bachmann IM, Brekke RL, Svendsen HL, Guttormsen AB, Aziz S, Dillekås H, Straume O. Toxic epidermal necrolysis after immune checkpoint inhibition, case report, and review of the literature. Acta Oncol 2022; 61:1295-1299. [PMID: 36073292 DOI: 10.1080/0284186x.2022.2119099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
- Hanne Sommerfelt
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Lene F Sandvik
- Department of Dermatology, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Ingeborg M Bachmann
- Department of Dermatology, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Ragnvald Ljones Brekke
- Department of Plastic, Hand, and Reconstructive surgery, National Burn Centre, Haukeland University Hospital, Bergen, Norway
| | - Henrik Løvendahl Svendsen
- Department of Plastic, Hand, and Reconstructive surgery, National Burn Centre, Haukeland University Hospital, Bergen, Norway
| | - Anne Berit Guttormsen
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Anaesthesia and Intensive Care, Haukeland University Hospital, Bergen, Norway
| | - Sura Aziz
- Department of Pathology, Haukeland University Hospital Bergen, Bergen, Norway
| | - Hanna Dillekås
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Oddbjørn Straume
- Department of Oncology, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway.,Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| |
Collapse
|
63
|
Che H, Xiong Q, Ma J, Chen S, Wu H, Xu H, Hou B. Association of Helicobacter pylori infection with survival outcomes in advanced gastric cancer patients treated with immune checkpoint inhibitors. BMC Cancer 2022; 22:904. [PMID: 35986342 PMCID: PMC9389789 DOI: 10.1186/s12885-022-10004-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 08/12/2022] [Indexed: 12/24/2022] Open
Abstract
Abstract
Background
Accumulating evidence has revealed that the gut microbiota influences the effectiveness of immune checkpoint inhibitors (ICIs) in cancer patients. As a part of the human microbiome, Helicobacter pylori (H. pylori) was reported to be associated with reduced effectiveness of anti-PD1 immunotherapy in patients with non-small-cell lung cancer (NSCLC). Gastric cancer is more closely related to H. pylori, so we conducted a retrospective analysis to verify whether the association of H. pylori and effectiveness is applicable to advanced gastric cancer (AGC) patients.
Material and methods
AGC patients who had evidence of H. pylori and received anti-PD-1 antibodies were enrolled in the study. The differences in the disease control rate (DCR), overall survival (OS) and progression-free survival (PFS) between the H. pylori-positive group and the negative group were compared.
Results
A total of 77 patients were included in this study; 34 patients were H. pylori positive, and the prevalence of H. pylori infection was 44.2%. Compared with the H. pylori-negative group, patients in the H. pylori-positive group had a higher risk of nonclinical response to anti-PD-1 antibody, with an OR of 2.91 (95% CI: 1.13–7.50). Patients in the H. pylori-negative group had a longer OS and PFS than those in the positive group, with an estimated median OS of 17.5 months vs. 6.2 months (HR = 2.85, 95% CI: 1.70–4.78; P = 0.021) and a median PFS of 8.4 months vs. 2.7 months (HR = 3.11, 95% CI: 1.96–5.07, P = 0.008). Multivariate analysis indicated that H. pylori infection was independently associated with PFS (HR = 1.90, 95% CI: 1.10–3.30; P = 0.022).
Conclusion
Our study unveils for the first time that H. pylori infection is associated with the outcome of immunotherapy for AGC patients. Multicenter, large sample and prospective clinical studies are needed to verify the association.
Collapse
|
64
|
Zhou J, Huang G, Wong WC, Hu DH, Zhu JW, Li R, Zhou H. The impact of antibiotic use on clinical features and survival outcomes of cancer patients treated with immune checkpoint inhibitors. Front Immunol 2022; 13:968729. [PMID: 35967438 PMCID: PMC9367677 DOI: 10.3389/fimmu.2022.968729] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 06/27/2022] [Indexed: 11/17/2022] Open
Abstract
Background Nowadays, immune checkpoint inhibitors (ICIs) have become one of the essential immunotherapies for cancer patients. However, the impact of antibiotic (ATB) use on cancer patients treated with ICIs remains controversial. Methods Our research included retrospective studies and a randomized clinical trial (RCT) with cancer patients treated with ICIs and ATB, from the public database of PubMed, Web of Science, Embase, Cochrane, clinical trials, and JAMA. The survival outcomes included progression-free survival (PFS) and overall survival (OS). Meanwhile, hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated, and subgroup analyses were performed to determine the concrete association between ATB use and the prognosis of cancer patients treated in ICIs. Results Our results revealed that ATB use was associated with poor survival outcomes, including OS (HR: 1.94, 95% CI: 1.68–2.25, p <0.001) and PFS (HR: 1.83, 95% CI: 1.53–2.19, p <0.001). The subgroup analysis learned about the association between ATB use and the prognosis of cancer patients with ICI treatment, including 5 cancer types, 3 kinds of ICI, 5 different ATP windows, broad-spectrum ATB class, and ECOG score. ATB treatment was associated with poor OS of non-small-cell lung cancer (NSCLC), renal cell carcinoma (RCC), esophageal cancer (EC), and melanoma (MEL) in patients treated in ICIs, while non-small-cell lung cancer (NSCLC) and renal cell carcinoma (RCC) were associated with poor PFS. Meanwhile, it was strongly related to the ICI type and ATB window. Furthermore, it is firstly mentioned that the use of broad-spectrum ATB class was strongly associated with poor PFS. Conclusion In conclusion, our meta-analysis indicated that ATB use was significantly associated with poor OS and PFS of cancer patients treated with ICI immunotherapy, especially for patients with ATB use in the period of (−60 days; +30 days) near the initiation of ICI treatment. Also, different cancer types and the ICI type can also impact the survival outcome. This first reveals the strong relationship between the broad-spectrum ATB class and poor PFS. Still, more studies are needed for further study.
Collapse
Affiliation(s)
- Jiaxin Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- International School, Jinan University, Guangzhou, China
| | - Guowei Huang
- Shunde Hospital Affiliated to Jinan University, Guangzhou, China
| | - Wan-Ching Wong
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Da-hai Hu
- International School, Jinan University, Guangzhou, China
| | - Jie-wen Zhu
- College of Science and Engineering, Jinan University, Guangzhou, China
| | - Ruiman Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- *Correspondence: Ruiman Li, ; Hong Zhou,
| | - Hong Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- *Correspondence: Ruiman Li, ; Hong Zhou,
| |
Collapse
|
65
|
Makaranka S, Scutt F, Frixou M, Wensley KE, Sharma R, Greenhowe J. The gut microbiome and melanoma: A review. Exp Dermatol 2022; 31:1292-1301. [PMID: 35793428 DOI: 10.1111/exd.14639] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/21/2022] [Accepted: 07/03/2022] [Indexed: 12/16/2022]
Abstract
Disturbances in the microbial ecosystem have been implemented in chronic inflammation, immune evasion and carcinogenesis, with certain microbes associated with the development of specific cancers. In recent times, the gut microbiome has been recognised as a potential novel player in the pathogenesis and treatment of malignant melanoma. It has been shown that the composition of gut microbiota in early-stage melanoma changes from in situ to invasive and then to metastatic disease. The gut bacterial and fungal profile has also been found to be significantly different in melanoma patients compared to controls. Multiple studies of immune checkpoint inhibitor (ICI) therapies have shown that the commensal microbiota may have an impact on anti-tumor immunity and therefore ICI response in cancer patients. When it comes to chemotherapy and radiotherapy treatments, studies demonstrate that gut microbiota are invaluable in the repair of radiation and chemotherapy-induced damage and therapeutic manipulation of gut microbiota can be an effective strategy to deal with side effects. Studies demonstrate the oncogenic and tumor-suppressive properties of the gut microbiome, which may play a role in the pathogenesis of melanoma. Despite this, investigations into specific interactions are still in its infancy, but starting to gain momentum as more significant and clinically relevant effects are emerging.
Collapse
Affiliation(s)
| | - Freya Scutt
- Department of Plastic Surgery, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Mikaela Frixou
- Department of Gastroenterology, Aberdeen Royal Infirmary, Aberdeen, UK
| | | | - Ravi Sharma
- Department of Oncology, Aberdeen Royal Infirmary, Aberdeen, UK
| | | |
Collapse
|
66
|
Statin use improves the efficacy of nivolumab in patients with advanced renal cell carcinoma. Eur J Cancer 2022; 172:191-198. [PMID: 35780525 DOI: 10.1016/j.ejca.2022.04.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 04/10/2022] [Accepted: 04/24/2022] [Indexed: 01/01/2023]
Abstract
BACKGROUND Statins are widely used in an ageing population, including subjects with solid malignancies. However, no conclusive evidence is currently available on their potential influence on patients' outcome. We aimed to assess whether statin exposure affects the survival of patients with metastatic renal cell carcinoma (mRCC) treated with nivolumab. PATIENTS AND METHODS Medical records of patients with documented mRCC treated with second- or third-line nivolumab were reviewed at ten institutions from Italy, Spain and the USA. Patients were assessed for overall survival (OS), progression-free survival (PFS), and overall clinical benefit. Univariate and multivariate analyses were used to explore the association of variables of interest with survival. RESULTS A total of 219 patients with mRCC receiving nivolumab between January 2016 and September 2021 were eligible for inclusion in this study; 59 (27%) were statin users. The median OS (34.4 versus 18.6 months, p = 0.017) and PFS (11.7 versus 4.6 months, p = 0.013) resulted apparently longer in statin users. Stratified by age, longer median OS and PFS were associated with statin exposure in both patients aged ≥70 y (median OS: 21.4 versus 10.1 months, p = 0.047; median PFS: 16.4 versus 4.6 months, p = 0.022) and <70 y (median OS: 34.4 versus 21.4 months, p = 0.043; median PFS: 10.3 versus 4.6 months, p = 0.042). Overall clinical benefit resulted higher in statin users than non-users (71% versus 54%, p = 0.030). CONCLUSIONS Our study suggests a prognostic impact of statin use in patients receiving nivolumab for mRCC.
Collapse
|
67
|
The ability of inflammatory markers to recognize infection in cancer patients with fever at admission. Immunol Res 2022; 70:667-677. [PMID: 35764901 DOI: 10.1007/s12026-022-09299-4] [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: 02/24/2022] [Accepted: 05/23/2022] [Indexed: 11/05/2022]
Abstract
Infection is one of the main causes of death in cancer patients. Accurate identification of fever caused by infection could avoid unnecessary antibiotic treatment and hospitalization. This study evaluated the diagnostic value of procalcitonin (PCT), C-reactive protein (CRP), interleukin-6 (IL-6), interleukin-10 (IL-10), and other commonly used inflammatory markers in suspected infected adult cancer patients with fever, for better use of antibiotics. This research retrospective analyzed the clinical data of 102 adult cancer patients with fever and compared the serum levels of commonly used inflammatory markers for different fever reasons. Receiver-operating characteristic (ROC) curve and logistic regression analyses were performed. In adult cancer patients with fever, the serum PCT, CRP, IL-6, and IL-10 levels of infected patients were significantly higher than uninfected patients (median 1.19 ng/ml vs 0.14 ng/ml, 93.11 mg/l vs 56.55 mg/l, 123.74 pg/ml vs 47.35 pg/ml, 8.74 pg/ml vs 3.22 pg/ml; Mann-Whitney p = 0.000, p = 0.009, p = 0.004, p = 0.000, respectively). The ROC area under the curve(AUC) was 0.769 (95% confidence interval (CI) 0.681-0.857; p = 0.000) for PCT, 0.664 (95% CI 0.554-0.775; p = 0.009) for CRP, 0.681(95% CI 0.576-0.785; p = 0.004) for IL-6, and 0.731(95% CI 0.627-0.834; p = 0.000) for IL-10. PCT had specificity of 96.67% and positive predictive value (PPV) of 97.6%, when the cut-off value is set as 0.69 ng/ml. The serum IL-6 and IL-10 levels also had significant differences between the infected and uninfected cancer patients with advanced disease (median 128.92 pg/ml vs 36.40 pg/ml, 8.05 pg/ml vs 2.92 pg/ml; Mann-Whitney p = 0.003, p = 0.001, respectively). For the patients with neutropenia, IL-6 and IL-10 had higher AUC of 0.811 and 0.928, respectively. With a cut-off of 9.10 pg/ml, IL-10 had the highest sensitivity 83.33% and specificity 100%. In adult cancer patients, PCT had the best performance compared to CRP, IL-6, and IL-10 in differentiating infected from uninfected causes of fever, with high specificity and PPV. IL-6 and IL-10 might be useful in cancer patients with severe bloodstream infections and advanced disease. However, for patients with neutropenia, IL-10 might be more valuable than PCT in diagnosing infection.
Collapse
|
68
|
Xi Y, Liu F, Qiu B, Li Y, Xie X, Guo J, Wu L, Liang T, Wang D, Wang J, Chen M, Xue L, Ding Y, Zhang J, Wu Q, Liu H. Analysis of Gut Microbiota Signature and Microbe-Disease Progression Associations in Locally Advanced Non-Small Cell Lung Cancer Patients Treated With Concurrent Chemoradiotherapy. Front Cell Infect Microbiol 2022; 12:892401. [PMID: 35719339 PMCID: PMC9200620 DOI: 10.3389/fcimb.2022.892401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/04/2022] [Indexed: 11/17/2022] Open
Abstract
Purpose To evaluate the association of gut microbiome signature and disease progression in locally advanced non-small cell lung cancer (LA-NSCLC) patients treated with concurrent chemoradiotherapy (CCRT) by fecal metagenome analysis. Methods Metagenome-wide association studies on baseline fecal samples from 18 LA-NSCLC patients before CCRT and 13 controls from healthy first-degree relatives were performed. Among the 18 LA-NSCLC patients, six patients were defined as the long progression-free survival (long-PFS) group (PFS≥11 months) while another 12 were in the short-PFS group (PFS<11 months). Alpha diversity, taxonomic composition, and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional pathways were compared between groups. Results The Firmicutes/Bacteroidetes value of long-PFS group was higher than those of short-PFS (p=0.073) and healthy individual groups (p=0.009). Meanwhile, long-PFS group had significantly higher diversities in Fungi, Archaea, and Viruses than short-PFS group. The KEGG pathways overrepresented in short-PFS group included fructose and mannose metabolism (p=0.028), streptomycin biosynthesis (p=0.028), acarbose and validamycin biosynthesis (p=0.013), ribosome biogenesis in eukaryotes (p=0.035), biosynthesis of vancomycin group antibiotics (p=0.004), apoptosis-fly (p=0.044), and tetracycline biosynthesis (p=0.044), while those overrepresented in long-PFS group included fatty acid biosynthesis (p=0.035), fatty acid metabolism (p=0.008), vancomycin resistance (p=0.008), longevity regulating pathway-worm (p=0.028), type II diabetes mellitus (p=0.004), and viral carcinogenesis (p=0.003). Further analysis of antibiotic resistome demonstrated that the short-PFS group had a trend with more antibiotic resistance genes than healthy control (p=0.070) and long-PFS groups (p=0.218). The vancomycin resistance sequences were significantly enriched in the long-PFS group compared to the short-PFS group (p=0.006). Conclusions The baseline gut microbiome composition and functionality might be associated with PFS in LA-NSCLC treated with CCRT. The outcome of CCRT might be modulated through bacterial metabolic pathways. The antibiotic resistance genes might play a role in disease progression and provide potential information on the relationship between the use of antibiotics and treatment efficacy of CCRT in LA-NSCLC.
Collapse
Affiliation(s)
- Yu Xi
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - FangJie Liu
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat−sen University Cancer Center, Guangzhou, China
| | - Bo Qiu
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat−sen University Cancer Center, Guangzhou, China
| | - Ying Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - XinQiang Xie
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - JinYu Guo
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat−sen University Cancer Center, Guangzhou, China
| | - Lei Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - TingTing Liang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - DaQuan Wang
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat−sen University Cancer Center, Guangzhou, China
| | - Juan Wang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Moutong Chen
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Liang Xue
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Yu Ding
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Jumei Zhang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - QingPing Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
- *Correspondence: Hui Liu, ; QingPing Wu,
| | - Hui Liu
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat−sen University Cancer Center, Guangzhou, China
- *Correspondence: Hui Liu, ; QingPing Wu,
| |
Collapse
|
69
|
Johnston CW, Badran AH. Natural and engineered precision antibiotics in the context of resistance. Curr Opin Chem Biol 2022; 69:102160. [PMID: 35660248 DOI: 10.1016/j.cbpa.2022.102160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/18/2022] [Accepted: 04/22/2022] [Indexed: 12/14/2022]
Abstract
Antibiotics are essential weapons in our fight against infectious disease, yet the consequences of broad-spectrum antibiotic use on microbiome stability and pathogen resistance are prompting investigations into more selective alternatives. Echoing the advent of precision medicine in oncology, precision antibiotics with focused activities are emerging as a means of addressing infections without damaging microbiomes or incentivizing resistance. Historically, antibiotic design principles have been gleaned from Nature, and reinvestigation of overlooked antibacterials is now providing scaffolds and targets for the design of pathogen-specific drugs. In this perspective, we summarize the biosynthetic and antibacterial mechanisms used to access these activities, and discuss how such strategies may be co-opted through engineering approaches to afford precision antibiotics.
Collapse
Affiliation(s)
- Chad W Johnston
- Department of Pharmacology & Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Ahmed H Badran
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
70
|
Oster P, Vaillant L, McMillan B, Velin D. The Efficacy of Cancer Immunotherapies Is Compromised by Helicobacter pylori Infection. Front Immunol 2022; 13:899161. [PMID: 35677057 PMCID: PMC9168074 DOI: 10.3389/fimmu.2022.899161] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/26/2022] [Indexed: 12/12/2022] Open
Abstract
Helicobacter pylori infects the gastric mucosa of a large number of humans. Although asymptomatic in the vast majority of cases, H pylori infection can lead to the development of peptic ulcers gastric adenocarcinoma and mucosa-associated lymphoid tissue (MALT) lymphoma. Using a variety of mechanisms, H pylori locally suppresses the function of the host immune system to establish chronic infection. Systemic immunomodulation has been observed in both clinical and pre-clinical studies, which have demonstrated that H pylori infection is associated with reduced incidence of inflammatory diseases, such as asthma and Crohn’s disease. The introduction of immunotherapies in the arsenal of anti-cancer drugs has revealed a new facet of H pylori-induced immune suppression. In this review, we will describe the intimate interactions between H pylori and its host, and formulate hypothtyeses describing the detrimental impact of H pylori infection on the efficacy of cancer immunotherapies.
Collapse
|
71
|
Metronomic Chemotherapy in Prostate Cancer. J Clin Med 2022; 11:jcm11102853. [PMID: 35628979 PMCID: PMC9143236 DOI: 10.3390/jcm11102853] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 02/01/2023] Open
Abstract
Despite the significant expansion of the therapeutic armamentarium associated with the introduction of novel endocrine therapies, cytotoxic agents, radiopharmaceuticals, and PARP inhibitors, progression of metastatic castration-resistant prostate cancer (mCRPC) beyond treatment options remains the leading cause of death in advanced prostate cancer patients. Metronomic chemotherapy (MC) is an old concept of wise utilization of cytotoxic agents administered continuously and at low doses. The metronomic is unique due to its multidimensional mechanisms of action involving: (i) inhibition of cancer cell proliferation, (ii) inhibition of angiogenesis, (iii) mitigation of tumor-related immunosuppression, (iv) impairment of cancer stem cell functions, and (v) modulation of tumor and host microbiome. MC has been extensively studied in advanced prostate cancer before the advent of novel therapies, and its actual activity in contemporary, heavily pretreated mCRPC patients is unknown. We have conducted a prospective analysis of consecutive cases of mCRPC patients who failed all available standard therapies to find the optimal MC regimen for phase II studies. The metronomic combination of weekly paclitaxel 60 mg/m2 i.v. with capecitabine 1500 mg/d p.o. and cyclophosphamide 50 mg/d p.o. was selected as the preferred regimen for a planned phase II study in heavily pretreated mCRPC patients.
Collapse
|
72
|
Tian X, Mei T, Yu M, Li Y, Ao R, Gong Y. The impact of antibiotic selection and interval time among advanced non-small cell lung cancer patients receiving prior antibacterial treatment and first-line chemotherapy. Cancer Med 2022; 11:4849-4864. [PMID: 35543371 PMCID: PMC9761060 DOI: 10.1002/cam4.4815] [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: 02/06/2022] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND To determine whether antibiotic use before chemotherapy is associated with chemotherapy responses and patient outcomes among NSCLC patients and define the optimal interval between chemotherapy initiation and antibiotic treatment. MATERIALS AND METHODS One thousand four hundred and four advanced NSCLC patients receiving first-line platinum-based doublets therapy were retrospectively analyzed. Kaplan-Meier curve evaluated the impact of antibiotic use and type of antibiotics on the survival of patients. The factors affect the patient's prognosis were further confirmed by Cox regression. The optimal interval between antibiotic treatment and the initiation of chemotherapy was determined by the X-tile program. RESULTS NSCLC patients of 33.5% advanced underwent broad-spectrum antibiotic treatment prior to chemotherapy. In the chemotherapy only (Chemo) and chemotherapy plus antiangiogenesis (Chemo-angio) treatment groups, prior antibiotic treatment was associated with worse OS (Chemo: 13.8 vs. 17.6 months, p < 0.001; Chemo-angio:11.9 vs. 18.1 months, p = 0.012) and PFS (Chemo: 3.7 vs. 5.8 months, p < 0.001; Chemo-angio: 3.1 vs. 5.9 months, p < 0.001). Cox regression analysis revealed prior antibiotic administration as an independent predictor of OS and PFS (HR for PFS/OS: 1.925/1.452, both p < 0.001). Antibiotic usage duration (HR for PFS/OS: 1.030/1.036, p = 0.009/0.001) and type (PFS/OS: p < 0.001/p = 0.01) also showed significant association with patient prognosis, with calculated interval time cutoff values of 2, 4, and 2 days for fluoroquinolones, β-lactamase inhibitors, and cephalosporins, respectively. CONCLUSION Antibiotic use before first-line chemotherapy was associated with poor results in advanced NSCLC patients; treatment length and type being strongly correlated with patient outcomes. Appropriate prolongation of the time between two treatments may enhance patient survival. Further prospective research is however necessary.
Collapse
Affiliation(s)
- Xiaoman Tian
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduPR.China,Department of OncologyChengdu Jinniu District People's HospitalChengduPR.China
| | - Ting Mei
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduPR.China
| | - Min Yu
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduPR.China
| | - Yanying Li
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduPR.China
| | - Rui Ao
- Department of OncologyChengdu Jinniu District People's HospitalChengduPR.China,Department of OncologySichuan Provincial People's HospitalChengduPR.China
| | - Youling Gong
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduPR.China
| |
Collapse
|
73
|
Murray C, Galvan E, Ontiveros C, Deng Y, Bai H, Padron AS, Hinchee-Rodriguez K, Garcia MG, Kornepati A, Conejo-Garcia J, Curiel TJ. Pharmacologic Tumor PDL1 Depletion with Cefepime or Ceftazidime Promotes DNA Damage and Sensitivity to DNA-Damaging Agents. Int J Mol Sci 2022; 23:5129. [PMID: 35563520 PMCID: PMC9099860 DOI: 10.3390/ijms23095129] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 12/10/2022] Open
Abstract
The interaction between tumor surface-expressed PDL1 and immune cell PD1 for the evasion of antitumor immunity is well established and is targeted by FDA-approved anti-PDL1 and anti-PD1 antibodies. Nonetheless, recent studies highlight the immunopathogenicity of tumor-intrinsic PDL1 signals that can contribute to the resistance to targeted small molecules, cytotoxic chemotherapy, and αPD1 immunotherapy. As genetic PDL1 depletion is not currently clinically tractable, we screened FDA-approved drugs to identify those that significantly deplete tumor PDL1. Among the candidates, we identified the β-lactam cephalosporin antibiotic cefepime as a tumor PDL1-depleting drug (PDD) that increases tumor DNA damage and sensitivity to DNA-damaging agents in vitro in distinct aggressive mouse and human cancer lines, including glioblastoma multiforme, ovarian cancer, bladder cancer, and melanoma. Cefepime reduced tumor PDL1 post-translationally through ubiquitination, improved DNA-damaging-agent treatment efficacy in vivo in immune-deficient and -proficient mice, activated immunogenic tumor STING signals, and phenocopied specific genetic PDL1 depletion effects. The β-lactam ring and its antibiotic properties did not appear contributory to PDL1 depletion or to these treatment effects, and the related cephalosporin ceftazidime produced similar effects. Our findings highlight the rapidly translated potential for PDDs to inhibit tumor-intrinsic PDL1 signals and improve DNA-damaging agents and immunotherapy efficacy.
Collapse
Affiliation(s)
- Clare Murray
- Graduate School of Biomedical Science, University of Texas Health, San Antonio, TX 78229, USA; (C.M.); (C.O.); (M.G.G.); (A.K.)
| | - Eva Galvan
- UT Health Mays Cancer Center, University of Texas Health, San Antonio, TX 78229, USA;
- Department of Radiation Oncology, University of Texas Health, San Antonio, TX 78229, USA
| | - Carlos Ontiveros
- Graduate School of Biomedical Science, University of Texas Health, San Antonio, TX 78229, USA; (C.M.); (C.O.); (M.G.G.); (A.K.)
| | - Yilun Deng
- Department of Medicine, University of Texas Health, San Antonio, TX 78229, USA; (Y.D.); (H.B.); (A.S.P.); (K.H.-R.)
| | - Haiyan Bai
- Department of Medicine, University of Texas Health, San Antonio, TX 78229, USA; (Y.D.); (H.B.); (A.S.P.); (K.H.-R.)
| | - Alvaro Souto Padron
- Department of Medicine, University of Texas Health, San Antonio, TX 78229, USA; (Y.D.); (H.B.); (A.S.P.); (K.H.-R.)
| | - Kathryn Hinchee-Rodriguez
- Department of Medicine, University of Texas Health, San Antonio, TX 78229, USA; (Y.D.); (H.B.); (A.S.P.); (K.H.-R.)
| | - Myrna G. Garcia
- Graduate School of Biomedical Science, University of Texas Health, San Antonio, TX 78229, USA; (C.M.); (C.O.); (M.G.G.); (A.K.)
| | - Anand Kornepati
- Graduate School of Biomedical Science, University of Texas Health, San Antonio, TX 78229, USA; (C.M.); (C.O.); (M.G.G.); (A.K.)
| | - Jose Conejo-Garcia
- Department of Immunology, Moffitt Cancer Institute, Tampa, FL 33612, USA;
| | - Tyler J. Curiel
- Graduate School of Biomedical Science, University of Texas Health, San Antonio, TX 78229, USA; (C.M.); (C.O.); (M.G.G.); (A.K.)
- UT Health Mays Cancer Center, University of Texas Health, San Antonio, TX 78229, USA;
- Department of Medicine, University of Texas Health, San Antonio, TX 78229, USA; (Y.D.); (H.B.); (A.S.P.); (K.H.-R.)
| |
Collapse
|
74
|
Cheung KS, Chan EW, Tam A, Wong IOL, Seto WK, Hung IFN, Wong ICK, Leung WK. Association between antibiotic consumption and colon and rectal cancer development in older individuals: A territory-wide study. Cancer Med 2022; 11:3863-3872. [PMID: 35488387 PMCID: PMC9582694 DOI: 10.1002/cam4.4759] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/18/2022] [Accepted: 03/29/2022] [Indexed: 12/24/2022] Open
Abstract
Background Antibiotics may alter colorectal cancer (CRC) risk due to gut dysbiosis. We aimed to study the specific and temporal effects of various antibiotics on CRC development in older individuals. Methods This was a territory‐wide retrospective cohort study. Subjects aged 60 years and older who did not have CRC diagnosed on screening/diagnostic colonoscopy diagnosed between 2005 and 2013 were recruited. Exclusion criteria were history of CRC, colectomy, inflammatory bowel disease, and CRC diagnosed within 6 months of index colonoscopy. Exposure was use of any antibiotics up to 5 years before colonoscopy. The primary outcomes were CRC diagnosed >6 m after colonoscopy. Covariates were patient demographics, history of colonic polyps/polypectomy, concomitant medication use (NSAIDs, COX‐2 inhibitors, aspirin, and statins), and performance of endoscopy centers (colonoscopy volume and polypectomy rate). Stratified analysis was conducted according to nature of antibiotics and location of cancer. Results Ninety seven thousand one hundred and sixty‐two eligible subjects (with 1026 [1.0%] cases of CRC) were identified, 58,704 (60.4%) of whom were exposed to antibiotics before index colonoscopy. Use of antibiotics was associated with a lower risk of cancer in rectum (adjusted hazard ratio [aHR]: 0.64, 95% CI: 0.54–0.76), but a higher risk of cancer in proximal colon (aHR: 1.63, 95%CI: 1.15–2.32). These effects differed as regards the anti‐anaerobic/anti‐aerobic activity, narrow‐/broad‐spectrum, and administration route of antibiotics. Conclusions Antibiotics had divergent effects on CRC development in older subjects, which varied according to the location of cancer, antibiotic class, and administration route.
Collapse
Affiliation(s)
- Ka Shing Cheung
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong City, Hong Kong.,Department of Medicine, The University of Hong Kong&Shenzhen Hospital, Shenzhen, China
| | - Esther W Chan
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong City, Hong Kong
| | - Anthony Tam
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong City, Hong Kong
| | - Irene O L Wong
- School of Public Health, The University of Hong Kong, Hong Kong City, Hong Kong
| | - Wai Kay Seto
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong City, Hong Kong.,Department of Medicine, The University of Hong Kong&Shenzhen Hospital, Shenzhen, China
| | - Ivan F N Hung
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong City, Hong Kong
| | - Ian C K Wong
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong City, Hong Kong.,UCL School of Pharmacy, University College London, London, UK
| | - Wai K Leung
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong City, Hong Kong
| |
Collapse
|
75
|
Morrell S, Kohonen-Corish MRJ, Ward RL, Sorrell TC, Roder D, Currow DC. Antibiotic exposure six months preceding systemic therapy was associated with lower cancer survival. J Clin Epidemiol 2022; 147:122-131. [DOI: 10.1016/j.jclinepi.2022.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 03/29/2022] [Accepted: 04/04/2022] [Indexed: 11/29/2022]
|
76
|
Impact of Baseline Clinical Biomarkers on Treatment Outcomes in Patients with Advanced NSCLC Receiving First-line Pembrolizumab-based Therapy. Clin Lung Cancer 2022; 23:438-445. [DOI: 10.1016/j.cllc.2022.03.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/28/2022] [Indexed: 12/29/2022]
|
77
|
Abstract
Accumulating evidence demonstrates that the oral pathobiont Fusobacterium nucleatum is involved in the progression of an increasing number of tumors types. Thus far, the mechanisms underlying tumor exacerbation by F. nucleatum include the enhancement of proliferation, establishment of a tumor‐promoting immune environment, induction of chemoresistance, and the activation of immune checkpoints. This review focuses on the mechanisms that mediate tumor‐specific colonization by fusobacteria. Elucidating the mechanisms mediating fusobacterial tumor tropism and promotion might provide new insights for the development of novel approaches for tumor detection and treatment.
Collapse
Affiliation(s)
- Tamar Alon-Maimon
- The Institute of Dental Sciences, The Hebrew University-Hadassah School of Dental Medicine, Jerusalem, Israel
| | - Ofer Mandelboim
- The Concern Foundation Laboratories, Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Faculty of Medicine, The Hebrew University Medical School, Jerusalem, Israel
| | - Gilad Bachrach
- The Institute of Dental Sciences, The Hebrew University-Hadassah School of Dental Medicine, Jerusalem, Israel
| |
Collapse
|
78
|
Poonacha KNT, Villa TG, Notario V. The Interplay among Radiation Therapy, Antibiotics and the Microbiota: Impact on Cancer Treatment Outcomes. Antibiotics (Basel) 2022; 11:331. [PMID: 35326794 PMCID: PMC8944497 DOI: 10.3390/antibiotics11030331] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/27/2022] [Accepted: 02/28/2022] [Indexed: 12/01/2022] Open
Abstract
Radiation therapy has been used for more than a century, either alone or in combination with other therapeutic modalities, to treat most types of cancer. On average, radiation therapy is included in the treatment plans for over 50% of all cancer patients, and it is estimated to contribute to about 40% of curative protocols, a success rate that may reach 90%, or higher, for certain tumor types, particularly on patients diagnosed at early disease stages. A growing body of research provides solid support for the existence of bidirectional interaction between radiation exposure and the human microbiota. Radiation treatment causes quantitative and qualitative changes in the gut microbiota composition, often leading to an increased abundance of potentially hazardous or pathogenic microbes and a concomitant decrease in commensal bacteria. In turn, the resulting dysbiotic microbiota becomes an important contributor to worsen the adverse events caused in patients by the inflammatory process triggered by the radiation treatment and a significant determinant of the radiation therapy anti-tumor effectiveness. Antibiotics, which are frequently included as prophylactic agents in cancer treatment protocols to prevent patient infections, may affect the radiation/microbiota interaction through mechanisms involving both their antimicrobial activity, as a mediator of microbiota imbalances, and their dual capacity to act as pro- or anti-tumorigenic effectors and, consequently, as critical determinants of radiation therapy outcomes. In this scenario, it becomes important to introduce the use of probiotics and/or other agents that may stabilize the healthy microbiota before patients are exposed to radiation. Ultimately, newly developed methodologies may facilitate performing personalized microbiota screenings on patients before radiation therapy as an accurate way to identify which antibiotics may be used, if needed, and to inform the overall treatment planning. This review examines currently available data on these issues from the perspective of improving radiation therapy outcomes.
Collapse
Affiliation(s)
| | - Tomás G. Villa
- Department of Microbiology, Faculty of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15705 La Coruña, Spain;
| | - Vicente Notario
- Department of Radiation Medicine, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
79
|
Oster P, Vaillant L, Riva E, McMillan B, Begka C, Truntzer C, Richard C, Leblond MM, Messaoudene M, Machremi E, Limagne E, Ghiringhelli F, Routy B, Verdeil G, Velin D. Helicobacter pylori infection has a detrimental impact on the efficacy of cancer immunotherapies. Gut 2022; 71:457-466. [PMID: 34253574 PMCID: PMC8862014 DOI: 10.1136/gutjnl-2020-323392] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 06/24/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE In this study, we determined whether Helicobacter pylori (H. pylori) infection dampens the efficacy of cancer immunotherapies. DESIGN Using mouse models, we evaluated whether immune checkpoint inhibitors or vaccine-based immunotherapies are effective in reducing tumour volumes of H. pylori-infected mice. In humans, we evaluated the correlation between H. pylori seropositivity and the efficacy of the programmed cell death protein 1 (PD-1) blockade therapy in patients with non-small-cell lung cancer (NSCLC). RESULTS In mice engrafted with MC38 colon adenocarcinoma or B16-OVA melanoma cells, the tumour volumes of non-infected mice undergoing anticytotoxic T-lymphocyte-associated protein 4 and/or programmed death ligand 1 or anti-cancer vaccine treatments were significantly smaller than those of infected mice. We observed a decreased number and activation status of tumour-specific CD8+ T cells in the tumours of infected mice treated with cancer immunotherapies independent of the gut microbiome composition. Additionally, by performing an in vitro co-culture assay, we observed that dendritic cells of infected mice promote lower tumour-specific CD8+ T cell proliferation. We performed retrospective human clinical studies in two independent cohorts. In the Dijon cohort, H. pylori seropositivity was found to be associated with a decreased NSCLC patient survival on anti-PD-1 therapy. The survival median for H. pylori seropositive patients was 6.7 months compared with 15.4 months for seronegative patients (p=0.001). Additionally, in the Montreal cohort, H. pylori seropositivity was found to be associated with an apparent decrease of NSCLC patient progression-free survival on anti-PD-1 therapy. CONCLUSION Our study unveils for the first time that the stomach microbiota affects the response to cancer immunotherapies and that H. pylori serology would be a powerful tool to personalize cancer immunotherapy treatment.
Collapse
Affiliation(s)
- Paul Oster
- Service of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Laurie Vaillant
- Service of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Erika Riva
- Service of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Brynn McMillan
- Service of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Christina Begka
- Service of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Caroline Truntzer
- Department of Medical Oncology, Centre Georges François Leclerc, Dijon, France
| | - Corentin Richard
- Research Centre for the University of Montréal (CRCHUM), Hematology-Oncology Division, Department of Medicine, University of Montreal Healthcare Centre (CHUM), Montreal, Quebec, Canada
| | - Marine M Leblond
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Meriem Messaoudene
- Research Centre for the University of Montréal (CRCHUM), Hematology-Oncology Division, Department of Medicine, University of Montreal Healthcare Centre (CHUM), Montreal, Quebec, Canada
| | - Elisavet Machremi
- Service of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Emeric Limagne
- Department of Medical Oncology, Centre Georges François Leclerc, Dijon, France
| | | | - Bertrand Routy
- Research Centre for the University of Montréal (CRCHUM), Hematology-Oncology Division, Department of Medicine, University of Montreal Healthcare Centre (CHUM), Montreal, Quebec, Canada
| | - Gregory Verdeil
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Dominique Velin
- Service of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
80
|
Derosa L, Routy B, Thomas AM, Iebba V, Zalcman G, Friard S, Mazieres J, Audigier-Valette C, Moro-Sibilot D, Goldwasser F, Silva CAC, Terrisse S, Bonvalet M, Scherpereel A, Pegliasco H, Richard C, Ghiringhelli F, Elkrief A, Desilets A, Blanc-Durand F, Cumbo F, Blanco A, Boidot R, Chevrier S, Daillère R, Kroemer G, Alla L, Pons N, Le Chatelier E, Galleron N, Roume H, Dubuisson A, Bouchard N, Messaoudene M, Drubay D, Deutsch E, Barlesi F, Planchard D, Segata N, Martinez S, Zitvogel L, Soria JC, Besse B. Intestinal Akkermansia muciniphila predicts clinical response to PD-1 blockade in patients with advanced non-small-cell lung cancer. Nat Med 2022; 28:315-324. [PMID: 35115705 DOI: 10.1038/s41591-021-01655-5] [Citation(s) in RCA: 276] [Impact Index Per Article: 138.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 12/06/2021] [Indexed: 12/13/2022]
Abstract
Aside from PD-L1 expression, biomarkers of response to immune checkpoint inhibitors (ICIs) in non-small-cell lung cancer (NSCLC) are needed. In a previous retrospective analysis, we documented that fecal Akkermansia muciniphila (Akk) was associated with clinical benefit of ICI in patients with NSCLC or kidney cancer. In the current study, we performed shotgun-metagenomics-based microbiome profiling in a large cohort of patients with advanced NSCLC (n = 338) treated with first- or second-line ICIs to prospectively validate the predictive value of fecal Akk. Baseline stool Akk was associated with increased objective response rates and overall survival in multivariate analyses, independent of PD-L1 expression, antibiotics, and performance status. Intestinal Akk was accompanied by a richer commensalism, including Eubacterium hallii and Bifidobacterium adolescentis, and a more inflamed tumor microenvironment in a subset of patients. However, antibiotic use (20% of cases) coincided with a relative dominance of Akk above 4.8% accompanied with the genus Clostridium, both associated with resistance to ICI. Our study shows significant differences in relative abundance of Akk that may represent potential biomarkers to refine patient stratification in future studies.
Collapse
Affiliation(s)
- Lisa Derosa
- Gustave Roussy Cancer Campus, Villejuif, France.,Cancer Medicine Department, Gustave Roussy, Villejuif, France.,Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée, Ligue Nationale contre le Cancer, Villejuif, France.,Université Paris-Saclay, Ile-de-France, France
| | - Bertrand Routy
- Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM), Hematology-Oncology Division, Montréal, Quebec, Canada.,Centre de Recherche du CHUM (CRCHUM), Montréal, Quebec, Canada
| | - Andrew Maltez Thomas
- Department CIBIO, University of Trento, Trento, Italy.,European Institute of Oncology (IEO) IRCCS, Milan, Italy
| | - Valerio Iebba
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Gerard Zalcman
- Thoracic Oncology Department-CIC1425/CLIP2 Paris-Nord, Hospital Bichat-Claude Bernard, AP-HP, Université Paris-Diderot, Paris, France
| | - Sylvie Friard
- Pneumology Department, Foch Hospital, Suresnes, France
| | - Julien Mazieres
- Department of Pneumology, Toulouse University Hospital, Toulouse, France
| | | | - Denis Moro-Sibilot
- Department of Thoracic Oncology, Centre Hospitalier Universitaire, Grenoble, France
| | - François Goldwasser
- UPR 4466, Paris Descartes University, Sorbonne Paris Cité, Paris, France.,Department of Medical Oncology, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.,Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Paris, France
| | - Carolina Alves Costa Silva
- Gustave Roussy Cancer Campus, Villejuif, France.,Cancer Medicine Department, Gustave Roussy, Villejuif, France
| | | | | | - Arnaud Scherpereel
- Department of Pulmonary and Thoracic Oncology, University of Lille, University Hospital (CHU), Lille, France
| | | | - Corentin Richard
- Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM), Hematology-Oncology Division, Montréal, Quebec, Canada.,Centre de Recherche du CHUM (CRCHUM), Montréal, Quebec, Canada
| | - François Ghiringhelli
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France.,Centre de Recherche INSERM LNC-UMR1231, Dijon, France.,Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | - Arielle Elkrief
- Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM), Hematology-Oncology Division, Montréal, Quebec, Canada.,Centre de Recherche du CHUM (CRCHUM), Montréal, Quebec, Canada
| | - Antoine Desilets
- Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM), Hematology-Oncology Division, Montréal, Quebec, Canada.,Centre de Recherche du CHUM (CRCHUM), Montréal, Quebec, Canada
| | | | - Fabio Cumbo
- Department CIBIO, University of Trento, Trento, Italy
| | - Aitor Blanco
- Department CIBIO, University of Trento, Trento, Italy
| | - Romain Boidot
- Unit of Molecular Biology, Department of Biology and Pathology of Tumors, Georges-François Leclerc Cancer Center, UNICANCER, Dijon, France
| | - Sandy Chevrier
- Unit of Molecular Biology, Department of Biology and Pathology of Tumors, Georges-François Leclerc Cancer Center, UNICANCER, Dijon, France
| | | | - Guido Kroemer
- Gustave Roussy Cancer Campus, Villejuif, France.,UPR 4466, Paris Descartes University, Sorbonne Paris Cité, Paris, France.,Centre de Recherche des Cordeliers, INSERM U1138, Equipe labellisée-Ligue contre le cancer, Université de Paris, Institut Universitaire de France, Paris, France.,Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Laurie Alla
- Université Paris-Saclay, INRAE, MGP, Jouy en Josas, France
| | - Nicolas Pons
- Université Paris-Saclay, INRAE, MGP, Jouy en Josas, France
| | | | | | - Hugo Roume
- Université Paris-Saclay, INRAE, MGP, Jouy en Josas, France
| | | | - Nicole Bouchard
- Centre Hospitalier de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Meriem Messaoudene
- Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM), Hematology-Oncology Division, Montréal, Quebec, Canada.,Centre de Recherche du CHUM (CRCHUM), Montréal, Quebec, Canada
| | | | - Eric Deutsch
- Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris-Saclay, Ile-de-France, France.,Department of Radiation Oncology, Gustave Roussy, Villejuif, France.,INSERM U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Villejuif, France
| | - Fabrice Barlesi
- Gustave Roussy Cancer Campus, Villejuif, France.,Cancer Medicine Department, Gustave Roussy, Villejuif, France
| | - David Planchard
- Gustave Roussy Cancer Campus, Villejuif, France.,Cancer Medicine Department, Gustave Roussy, Villejuif, France
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy.,European Institute of Oncology (IEO) IRCCS, Milan, Italy
| | - Stéphanie Martinez
- Service des Maladies Respiratoires, Centre Hospitalier d'Aix-en-Provence, Aix-en-Provence, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Villejuif, France. .,Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée, Ligue Nationale contre le Cancer, Villejuif, France. .,Université Paris-Saclay, Ile-de-France, France. .,Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS) 1428, Villejuif, France.
| | | | - Benjamin Besse
- Gustave Roussy Cancer Campus, Villejuif, France.,Cancer Medicine Department, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Ile-de-France, France
| |
Collapse
|
81
|
Efficacy of atezolizumab in patients with advanced non-small cell lung cancer receiving concomitant antibiotic or proton pump inhibitor treatment: pooled analysis of five randomised control trials. J Thorac Oncol 2022; 17:758-767. [DOI: 10.1016/j.jtho.2022.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/27/2022] [Accepted: 02/07/2022] [Indexed: 10/19/2022]
|
82
|
Newsome RC, Yang Y, Jobin C. The microbiome, gastrointestinal cancer, and immunotherapy. J Gastroenterol Hepatol 2022; 37:263-272. [PMID: 34820895 PMCID: PMC9922516 DOI: 10.1111/jgh.15742] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022]
Abstract
The gastrointestinal tract greatly contributes to global cancer burden and cancer-related deaths. The microbiota represents the population of microorganisms that live in and around the body, located primarily in the gastrointestinal tract. The microbiota has been implicated in colorectal cancer development and progression, but its role in cancer therapy for the gastrointestinal tract is less defined, especially for extra-intestinal cancers. In this review, we discuss the past 5 years of research into microbial involvement in immune-related therapies for colorectal, pancreatic, hepatic, and gastric cancers, with the goal of highlighting recent advances and new areas for investigation in this field.
Collapse
Affiliation(s)
- Rachel C Newsome
- Departments of Medicine, University of Florida, Gainesville, Florida, USA
| | - Ye Yang
- Departments of Medicine, University of Florida, Gainesville, Florida, USA
| | - Christian Jobin
- Departments of Medicine, University of Florida, Gainesville, Florida, USA
- Infectious Diseases and Immunology, University of Florida, Gainesville, Florida, USA
- Anatomy and Cell Biology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
83
|
Jing Y, Chen X, Li K, Liu Y, Zhang Z, Chen Y, Liu Y, Wang Y, Lin SH, Diao L, Wang J, Lou Y, Johnson DB, Chen X, Liu H, Han L. Association of antibiotic treatment with immune-related adverse events in patients with cancer receiving immunotherapy. J Immunother Cancer 2022; 10:jitc-2021-003779. [PMID: 35058327 PMCID: PMC8772460 DOI: 10.1136/jitc-2021-003779] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2021] [Indexed: 12/17/2022] Open
Abstract
Background To determine whether antibiotic treatment is a risk factor for immune-related adverse events (irAEs) across different patients with cancer receiving anti-PD-1/PD-L1 therapies. Methods The retrospective analysis includes clinical information from 767 patients with cancer treated at Hunan Cancer Hospital from 2017 to 2020. The pharmacovigilance data analysis includes individual cases of 38,705 safety reports from the US Food and Drug Administration Adverse Event Reporting System (FAERS) from 2014 to 2020, and 25,122 cases of safety reports from the World Health Organization database VigiBase from 2014 to 2019. All cases that received anti-PD-1/PD-L1 treatment were included. Multiomics data from patients across 25 cancer types were download from The Cancer Genome Atlas. Logistic regression and propensity score algorithm was employed to calculate OR of irAEs. Results Retrospective analysis of in-house patients showed that irAE potential risks are higher in all cancer (OR 2.12, 95% CI 1.38 to 3.22, false discovery rate (FDR) adjusted-p=1.93×10−3) and patients with lung cancer (OR 3.16, 95% CI 1.67 to 5.95, FDR adjusted-p=1.93×10−3) when using antibiotics. Potential risk of irAEs in patients with lung cancer with antibiotic treatment is significantly higher in FAERS (OR 1.39, 95% CI 1.21 to 1.59; FDR adjusted-p=1.62×10−5) and VigiBase (OR 1.32, 95% CI 1.09 to 1.59, FDR adjusted-p=0.05). Mechanistically, decreased microbial diversity caused by antibiotics use may increase the irAE risk through mediating the irAE-related factors. Conclusions Our study is the first to comprehensively demonstrate the associations of irAEs and antibiotic during anti-PD-1/PD-L1 therapy across a wide spectrum of cancers by analyzing multisource data. Administration of antibiotics should be carefully evaluated in patients with cancer treated by anti-PD-1/PD-L1 to avoid potentially increasing irAE risk.
Collapse
Affiliation(s)
- Ying Jing
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas, USA
| | - Xue Chen
- Early Clinical Trial Center, Office of National Drug Clinical Trial Institution, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Kunyan Li
- Early Clinical Trial Center, Office of National Drug Clinical Trial Institution, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yaoming Liu
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas, USA
| | - Zhao Zhang
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas, USA
| | - Yiqing Chen
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas, USA
| | - Yuan Liu
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas, USA
| | - Yushu Wang
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas, USA
| | - Steven H Lin
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lixia Diao
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jing Wang
- Early Clinical Trial Center, Office of National Drug Clinical Trial Institution, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yanyan Lou
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, Florida, USA
| | - Douglas B Johnson
- Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Xiang Chen
- Department of Dermatology, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Liu
- Department of Dermatology, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Leng Han
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas, USA
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas, USA
- Department of Translational Medical Sciences, College of Medicine, Texas A&M University, Houston, TX, USA
| |
Collapse
|
84
|
Li H, Xiao Y, Li Q, Yao J, Yuan X, Zhang Y, Yin X, Saito Y, Fan H, Li P, Kuo WL, Halpin A, Gibbons DL, Yagita H, Zhao Z, Pang D, Ren G, Yee C, Lee JJ, Yu D. The allergy mediator histamine confers resistance to immunotherapy in cancer patients via activation of the macrophage histamine receptor H1. Cancer Cell 2022; 40:36-52.e9. [PMID: 34822775 PMCID: PMC8779329 DOI: 10.1016/j.ccell.2021.11.002] [Citation(s) in RCA: 114] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 09/01/2021] [Accepted: 11/04/2021] [Indexed: 01/12/2023]
Abstract
Reinvigoration of antitumor immunity remains an unmet challenge. Our retrospective analyses revealed that cancer patients who took antihistamines during immunotherapy treatment had significantly improved survival. We uncovered that histamine and histamine receptor H1 (HRH1) are frequently increased in the tumor microenvironment and induce T cell dysfunction. Mechanistically, HRH1-activated macrophages polarize toward an M2-like immunosuppressive phenotype with increased expression of the immune checkpoint VISTA, rendering T cells dysfunctional. HRH1 knockout or antihistamine treatment reverted macrophage immunosuppression, revitalized T cell cytotoxic function, and restored immunotherapy response. Allergy, via the histamine-HRH1 axis, facilitated tumor growth and induced immunotherapy resistance in mice and humans. Importantly, cancer patients with low plasma histamine levels had a more than tripled objective response rate to anti-PD-1 treatment compared with patients with high plasma histamine. Altogether, pre-existing allergy or high histamine levels in cancer patients can dampen immunotherapy responses and warrant prospectively exploring antihistamines as adjuvant agents for combinatorial immunotherapy.
Collapse
Affiliation(s)
- Hongzhong Li
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yi Xiao
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Qin Li
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jun Yao
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiangliang Yuan
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yuan Zhang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xuedong Yin
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yohei Saito
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Huihui Fan
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ping Li
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wen-Ling Kuo
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Angela Halpin
- Enterprise Data Engineering & Analytics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Da Pang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Guosheng Ren
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China
| | - Cassian Yee
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - J Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
85
|
Tarantino P, Barroso-Sousa R, Garrido-Castro AC, McAllister SS, Guerriero JL, Mittendorf E, Curigliano G, Tolaney SM. Understanding resistance to immune checkpoint inhibitors in advanced breast cancer. Expert Rev Anticancer Ther 2021; 22:141-153. [PMID: 34919490 DOI: 10.1080/14737140.2022.2020650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION The addition of immune checkpoint inhibitors (ICIs) to frontline chemotherapy has improved survival for patients with advanced triple-negative breast cancer (TNBC) expressing programmed death-ligand 1 (PD-L1). Nonetheless, most patients develop resistance, with outcomes remaining poor for this population. Moreover, unsatisfactory activity has been observed with ICIs in PD-L1-negative TNBC and in other breast cancer (BC) subtypes, warranting a deeper understanding of resistance to ICIs in BC. AREAS COVERED We discuss the immune landscape of distinct BC subtypes, review the clinical activity of immunotherapy in BC, and highlight strategies under development to overcome resistance to ICIs. EXPERT OPINION Activity and resistance to ICIs in BC are strongly related to the intrinsic immunophenotype of the tumor tissue. Several promising biomarkers reflecting the immunological state of BC are emerging, with only PD-L1 expression currently adopted into clinical practice. However, limitations make of PD-L1 a sub-optimal biomarker for patient selection, which require efforts to integrate this marker with other immunological features. Concomitantly, a wide variety of drug combinations designed to overcome immune-resistance are being evaluated, with some encouraging signals observed in early-phase trials. Combination strategies tailored to patient and tumor immunophenotype may allow to overcome resistance and fully exploit the potential of ICIs.
Collapse
Affiliation(s)
- Paolo Tarantino
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.,Breast Oncology Program Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | | | - Ana C Garrido-Castro
- Breast Oncology Program Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Sandra S McAllister
- Harvard Medical School, Boston, MA, USA.,Hematology Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Jennifer L Guerriero
- Breast Oncology Program Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Elizabeth Mittendorf
- Breast Oncology Program Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Sara M Tolaney
- Breast Oncology Program Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
86
|
Kubeček O, Paterová P, Novosadová M. Risk Factors for Infections, Antibiotic Therapy, and Its Impact on Cancer Therapy Outcomes for Patients with Solid Tumors. Life (Basel) 2021; 11:1387. [PMID: 34947918 PMCID: PMC8705721 DOI: 10.3390/life11121387] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/05/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022] Open
Abstract
Infections represent a significant cause of morbidity and mortality in cancer patients. Multiple factors related to the patient, tumor, and cancer therapy can affect the risk of infection in patients with solid tumors. A thorough understanding of such factors can aid in the identification of patients with substantial risk of infection, allowing medical practitioners to tailor therapy and apply prophylactic measures to avoid serious complications. The use of novel treatment modalities, including targeted therapy and immunotherapy, brings diagnostic and therapeutic challenges into the management of infections in cancer patients. A growing body of evidence suggests that antibiotic therapy can modulate both toxicity and antitumor response induced by chemotherapy, radiotherapy, and especially immunotherapy. This article provides a comprehensive review of potential risk factors for infections and therapeutic approaches for the most prevalent infections in patients with solid tumors, and discusses the potential effect of antibiotic therapy on toxicity and efficacy of cancer therapy.
Collapse
Affiliation(s)
- Ondřej Kubeček
- Department of Oncology and Radiotherapy, Faculty of Medicine and University Hospital in Hradec Králové, Charles University, Sokolská 581, 50005 Hradec Králové, Czech Republic;
| | - Pavla Paterová
- Department of Clinical Microbiology, Faculty of Medicine and University Hospital in Hradec Králové, Charles University, Sokolská 581, 50005 Hradec Králové, Czech Republic
| | - Martina Novosadová
- Department of Clinical Pharmacy, Hospital Pharmacy, University Hospital in Hradec Králové, Sokolská 581, 50005 Hradec Králové, Czech Republic;
| |
Collapse
|
87
|
Klümper N, Schmucker P, Hahn O, Höh B, Mattigk A, Banek S, Ellinger J, Heinzelbecker J, Sikic D, Eckstein M, Strauß A, Zengerling F, Hölzel M, Zeuschner P, Kalogirou C. C‐reactive protein flare‐response predicts long‐term efficacy to first‐line anti‐PD‐1‐based combination therapy in metastatic renal cell carcinoma. Clin Transl Immunology 2021; 10:e1358. [PMID: 34925829 PMCID: PMC8648498 DOI: 10.1002/cti2.1358] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/01/2021] [Accepted: 11/17/2021] [Indexed: 11/17/2022] Open
Abstract
Objectives Immune checkpoint blockade (IO) has revolutionised the treatment of metastatic renal cell carcinoma (mRCC). Early C‐reactive protein (CRP) kinetics, especially the recently introduced CRP flare‐response phenomenon, has shown promising results to predict IO efficacy in mRCC, but has only been studied in second line or later. Here, we aimed to validate the predictive value of early CRP kinetics for 1st‐line treatment of mRCC with αPD‐1 plus either αCTLA‐4 (IO+IO) or tyrosine kinase inhibitor (IO+TKI). Methods In this multicentre retrospective study, we investigated the predictive potential of early CRP kinetics during 1st‐line IO therapy. Ninety‐five patients with mRCC from six tertiary referral centres with either IO+IO (N = 59) or IO+TKI (N = 36) were included. Patients were classified as CRP flare‐responders, CRP responders or non‐CRP responders as previously described, and their oncological outcome was compared. Results Our data validate the predictive potential of early CRP kinetics in 1st‐line immunotherapy in mRCC. CRP responders, especially CRP flare‐responders, had significantly prolonged progression‐free survival (PFS) compared with non‐CRP responders (median PFS: CRP flare‐responder: 19.2 months vs. responders: 16.2 vs. non‐CRP responders: 5.6, P < 0.001). In both the IO+IO and IO+TKI subgroups, early CRP kinetics remained significantly associated with improved PFS. CRP flare‐response was also associated with long‐term response ≥ 12 months. Conclusions Early CRP kinetics appears to be a low‐cost and easy‐to‐implement on‐treatment biomarker to predict response to 1st‐line IO combination therapy. It has potential to optimise therapy monitoring and might represent a new standard of care biomarker for immunotherapy in mRCC.
Collapse
Affiliation(s)
- Niklas Klümper
- Department of Urology and Paediatric Urology University Hospital Bonn (UKB) Bonn Germany
- Institute of Experimental Oncology University Hospital Bonn (UKB) Bonn Germany
| | - Philipp Schmucker
- Department of Urology and Paediatric Urology Julius Maximilians University Medical Center of Würzburg Würzburg Germany
| | - Oliver Hahn
- Department of Urology University Medical Center Göttingen Göttingen Germany
| | - Benedikt Höh
- Department of Urology University Hospital Frankfurt Goethe University Frankfurt am Main Frankfurt Germany
| | - Angelika Mattigk
- Department of Urology and Paediatric Urology University Hospital Ulm Ulm Germany
| | - Severine Banek
- Department of Urology University Hospital Frankfurt Goethe University Frankfurt am Main Frankfurt Germany
| | - Jörg Ellinger
- Department of Urology and Paediatric Urology University Hospital Bonn (UKB) Bonn Germany
| | - Julia Heinzelbecker
- Department of Urology and Paediatric Urology Saarland University Homburg/Saar Germany
| | - Danijel Sikic
- Department of Urology and Pediatric Urology University Hospital Erlangen Friedrich‐Alexander‐University Erlangen‐Nuremberg Erlangen Germany
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN) Erlangen Germany
| | - Markus Eckstein
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN) Erlangen Germany
- Institute of Pathology University Hospital Erlangen Friedrich‐Alexander‐University Erlangen‐Nuremberg Erlangen Germany
| | - Arne Strauß
- Department of Urology University Medical Center Göttingen Göttingen Germany
| | | | - Michael Hölzel
- Institute of Experimental Oncology University Hospital Bonn (UKB) Bonn Germany
| | - Philip Zeuschner
- Department of Urology and Paediatric Urology Saarland University Homburg/Saar Germany
| | - Charis Kalogirou
- Department of Urology and Paediatric Urology Julius Maximilians University Medical Center of Würzburg Würzburg Germany
| |
Collapse
|
88
|
Enhanced Bellmunt Risk Score for Survival Prediction in Urothelial Carcinoma Treated With Immunotherapy. Clin Genitourin Cancer 2021; 20:132-138. [PMID: 34953754 DOI: 10.1016/j.clgc.2021.11.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 11/20/2022]
Abstract
INTRODUCTION The discrimination performance of Bellmunt risk score for immune checkpoint inhibitor (ICI) therapy is largely unknown. This study aimed to validate and enhance discrimination of the Bellmunt score in patients with urothelial carcinoma treated with ICIs. PATIENTS AND METHODS Cox proportional hazard analysis were used to validate overall survival (OS) discrimination performance of the Bellmunt score in patients with urothelial carcinoma treated with atezolizumab in IMvigor210. The c-statistic (c) was used to evaluate the ability of C-reactive protein (CRP), neutrophil-to-lymphocyte ratio (NLR), lactate dehydrogenase (LDH), PD-L1 gene expression level on immune cells (PD-L1 ICs), albumin, time from prior chemotherapy, and tumor site count to enhance the Bellmunt score. External validation of an enhanced Bellmunt score utilized the independent atezolizumab arm of IMvigor211. RESULTS In IMvigor210, Bellmunt score displayed moderate OS discrimination (c = 0.66). Addition of CRP (one point for CRP>30 mg/L) to the Bellmunt score resulted in greatest improvement in performance (c = 0.70), followed by NLR (c = 0.69). On external validation, CRP-Bellmunt score had superior performance (OS c = 0.67, PFS c = 0.60) than original Bellmunt score (OS c = 0.64, PFS c = 0.59) with 30% of patients reclassified into a higher risk group. Patients with CRP-Bellmunt score of 0, 1, 2, or 3-plus had 1-year OS probabilities of 63%, 44%, 21%, and 15%, respectively. CONCLUSION CRP inclusion within the Bellmunt score enhanced the ability to discriminate high risk patients misclassified using the original model. We propose that the CRP-Bellmunt score may enable improved patient stratification in ICI clinical trials and provide more accurate prognostic information for patients with urothelial carcinoma initiating ICIs.
Collapse
|
89
|
Kovács T, Mikó E, Ujlaki G, Yousef H, Csontos V, Uray K, Bai P. The involvement of oncobiosis and bacterial metabolite signaling in metastasis formation in breast cancer. Cancer Metastasis Rev 2021; 40:1223-1249. [PMID: 34967927 PMCID: PMC8825384 DOI: 10.1007/s10555-021-10013-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/15/2021] [Indexed: 12/15/2022]
Abstract
Breast cancer, the most frequent cancer in women, is characterized by pathological changes to the microbiome of breast tissue, the tumor, the gut, and the urinary tract. Changes to the microbiome are determined by the stage, grade, origin (NST/lobular), and receptor status of the tumor. This year is the 50th anniversary of when Hill and colleagues first showed that changes to the gut microbiome can support breast cancer growth, namely that the oncobiome can reactivate excreted estrogens. The currently available human and murine data suggest that oncobiosis is not a cause of breast cancer, but can support its growth. Furthermore, preexisting dysbiosis and the predisposition to cancer are transplantable. The breast's and breast cancer's inherent microbiome and the gut microbiome promote breast cancer growth by reactivating estrogens, rearranging cancer cell metabolism, bringing about a more inflammatory microenvironment, and reducing the number of tumor-infiltrating lymphocytes. Furthermore, the gut microbiome can produce cytostatic metabolites, the production of which decreases or blunts breast cancer. The role of oncobiosis in the urinary tract is largely uncharted. Oncobiosis in breast cancer supports invasion, metastasis, and recurrence by supporting cellular movement, epithelial-to-mesenchymal transition, cancer stem cell function, and diapedesis. Finally, the oncobiome can modify the pharmacokinetics of chemotherapeutic drugs. The microbiome provides novel leverage on breast cancer that should be exploited for better management of the disease.
Collapse
Affiliation(s)
- Tünde Kovács
- Department Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Edit Mikó
- Department Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Gyula Ujlaki
- Department Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Heba Yousef
- Department Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Viktória Csontos
- Department Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Karen Uray
- Department Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Peter Bai
- Department Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary.
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, 4032, Hungary.
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary.
| |
Collapse
|
90
|
Bardoscia L, Pasinetti N, Triggiani L, Cozzi S, Sardaro A. Biological Bases of Immune-Related Adverse Events and Potential Crosslinks With Immunogenic Effects of Radiation. Front Pharmacol 2021; 12:746853. [PMID: 34790123 PMCID: PMC8591245 DOI: 10.3389/fphar.2021.746853] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 10/19/2021] [Indexed: 01/03/2023] Open
Abstract
Immune checkpoint inhibitors have gained an established role in the treatment of different tumors. Indeed, their use has dramatically changed the landscape of cancer care, especially for tumor types traditionally known to have poor outcomes. However, stimulating anticancer immune responses may also elicit an unusual pattern of immune-related adverse events (irAEs), different from those of conventional chemotherapy, likely due to a self-tolerance impairment featuring the production of autoreactive lymphocytes and autoantibodies, or a non-specific autoinflammatory reaction. Ionizing radiation has proven to promote both positive pro-inflammatory and immunostimolatory activities, and negative anti-inflammatory and immunosuppressive mechanisms, as a result of cross-linked interactions among radiation dose, the tumor microenvironment and the host genetic predisposition. Several publications argue in favor of combining immunotherapy and a broad range of radiation schedules, based on the recent evidence of superior treatment responses and patient survival. The synergistic modulation of the immune response by radiation therapy and immunotherapeutics, particularly those manipulating T-cell activation, may also affect the type and severity of irAEs, suggesting a relationship between the positive antitumor and adverse autoimmune effects of these agents. As yet, information on factors that may help to predict immune toxicity is still lacking. The aim of our work is to provide an overview of the biological mechanisms underlying irAEs and possible crosslinks with radiation-induced anticancer immune responses. We believe such an overview may support the optimization of immunotherapy and radiotherapy as essential components of multimodal anticancer therapeutic approaches. Challenges in translating these to clinical practice are discussed.
Collapse
Affiliation(s)
- Lilia Bardoscia
- Radiation Oncology Unit, S. Luca Hospital, Healthcare Company Tuscany Nord Ovest, Lucca, Italy
| | - Nadia Pasinetti
- Radiation Oncology Department, ASST Valcamonica Esine and University of Brescia, Brescia, Italy
| | - Luca Triggiani
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Salvatore Cozzi
- Radiotherapy Unit, Clinical Cancer Centre, AUSL-IRCCS, Reggio Emilia, Italy
| | - Angela Sardaro
- Interdisciplinary Department of Medicine, Section of Radiology and Radiation Oncology, University of Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
91
|
Giordan Q, Salleron J, Vallance C, Moriana C, Clement-Duchene C. Impact of Antibiotics and Proton Pump Inhibitors on Efficacy and Tolerance of Anti-PD-1 Immune Checkpoint Inhibitors. Front Immunol 2021; 12:716317. [PMID: 34777340 PMCID: PMC8578856 DOI: 10.3389/fimmu.2021.716317] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/04/2021] [Indexed: 12/19/2022] Open
Abstract
Background The use of antibiotics (ATB) and proton-pump inhibitors (PPI) alters the composition and diversity of the gut microbiota, which can influence the immune system, consequently interfering with response to anti-PD1 immune checkpoint inhibitors (ICI). We assessed the impact of ATB and/or PPI use on the efficacy and safety of ICI. Methods Two hundred twelve patients treated with anti-PD1 ICI for non-small cell lung carcinoma, melanoma, upper airway & digestive tract carcinoma or renal cell carcinoma were retrospectively included. Patients having received ATB within 60 days before ICI initiation were included in the ATB+ group. Patients having received PPI within 30 days before ICI initiation were included in the PPI+ group. Four groups were thus considered: ATB-/PPI-, ATB+/PPI-, ATB-/PPI+, ATB+/PPI+. Response rate was assessed by RECIST v1.1. Overall survival (OS), progression-free survival (PFS) and adverse events, recorded using Common Terminology Criteria for Adverse Events Version 5, were compared using inverse probability of treatment weighting to account for selection bias. Results PFS at 6 months was 56.7 %, 95%CI (49.6%; 63.2%) and 47.2 %, 95%CI (39.8%;54.1%) at 12 months. OS was 81.6%, 95%CI (75.6%; 86.2%) at 6 months, and 69.4%, 95%CI (61.9%;75.7%) at 12 months. Compared to ATB-/PPI- group, PFS was lower for the ATB+/PPI- group [Hazard ratio (HR) 1.90, 95%CI (1.41;2.57)] and the ATB-/PPI+ group [HR 1.51, 95%CI (1.11;2.05)], and lowest in the ATB+/PPI+ group [HR 3.65, 95%CI (2.75;4.84)]. For OS, the use of ATB alone or PPI alone or in combination was a risk factor for death, with each increasing HR values by a similar magnitude, and the combination of ATB and PPI did not increase risk further. AEs were observed in 78 cases (36.8%) with no significant impact of ATB or PPI use. Conclusions This study reveals that ATB and/or PPI use can alter response to anti-PD1 ICI, and the prognosis of cancer patients. The microbiota mechanisms involved in the response to ICI should be investigated to optimize patient management.
Collapse
Affiliation(s)
- Quentin Giordan
- Department of Pharmacy, Centre Hospitalier Régional de Metz-Thionville, Metz, France
| | - Julia Salleron
- Biostatistics Unit, Institut de Cancérologie de Lorraine, Vandœuvre-lès-Nancy, France
| | - Catherine Vallance
- Department of Pharmacy, Institut de Cancérologie de Lorraine, Vandœuvre-lès-Nancy, France
| | - Clothilde Moriana
- Department of Internal Medicine, Centre Hospitalier Régional Universitaire de Nancy, Vandœuvre-lès-Nancy, France
| | | |
Collapse
|
92
|
Yang P, Wang Z, Peng Q, Lian W, Chen D. Comparison of the Gut Microbiota in Patients with Benign and Malignant Breast Tumors: A Pilot Study. Evol Bioinform Online 2021; 17:11769343211057573. [PMID: 34795472 PMCID: PMC8593289 DOI: 10.1177/11769343211057573] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/15/2021] [Indexed: 12/18/2022] Open
Abstract
The microbiome plays diverse roles in many diseases and can potentially contribute to cancer development. Breast cancer is the most commonly diagnosed cancer in women worldwide. Thus, we investigated whether the gut microbiota differs between patients with breast carcinoma and those with benign tumors. The DNA of the fecal microbiota community was detected by Illumina sequencing and the taxonomy of 16S rRNA genes. The α-diversity and β-diversity analyses were used to determine richness and evenness of the gut microbiota. Gene function prediction of the microbiota in patients with benign and malignant carcinoma was performed using PICRUSt. There was no significant difference in the α-diversity between patients with benign and malignant tumors (P = 3.15e-1 for the Chao index and P = 3.1e-1 for the ACE index). The microbiota composition was different between the 2 groups, although no statistical difference was observed in β-diversity. Of the 31 different genera compared between the 2 groups, level of only Citrobacter was significantly higher in the malignant tumor group than that in benign tumor group. The metabolic pathways of the gut microbiome in the malignant tumor group were significantly different from those in benign tumor group. Furthermore, the study establishes the distinct richness of the gut microbiome in patients with breast cancer with different clinicopathological factors, including ER, PR, Ki-67 level, Her2 status, and tumor grade. These findings suggest that the gut microbiome may be useful for the diagnosis and treatment of malignant breast carcinoma.
Collapse
Affiliation(s)
- Peidong Yang
- Department of Breast Surgery, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Zhitang Wang
- Department of Breast Surgery, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Qingqin Peng
- Department of Radiation Oncology, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Weibin Lian
- Department of Breast Surgery, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Debo Chen
- Department of Breast Surgery, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
93
|
Amedei A, Capasso C, Nannini G, Supuran CT. Microbiota, Bacterial Carbonic Anhydrases, and Modulators of Their Activity: Links to Human Diseases? Mediators Inflamm 2021; 2021:6926082. [PMID: 34803517 PMCID: PMC8601860 DOI: 10.1155/2021/6926082] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
The involvement of the human microbiome is crucial for different host functions such as protection, metabolism, reproduction, and especially immunity. However, both endogenous and exogenous factors can affect the balance of the microbiota, creating a state of dysbiosis, which can start various gastrointestinal or systemic diseases. The challenge of future medicine is to remodel the intestinal microbiota to bring it back to healthy equilibrium (eubiosis) and, thus, counteract its negative role in the diseases' onset. The shaping of the microbiota is currently practiced in different ways ranging from diet (or use of prebiotics, probiotics, and synbiotics) to phage therapy and antibiotics, including microbiota fecal transplantation. Furthermore, because microbiota modulation is a capillary process, and because many microbiota bacteria (both beneficial and pathogenic) have carbonic anhydrases (specifically the four classes α, β, γ, and ι), we believe that the use of CA inhibitors and activators can open up new therapeutic strategies for many diseases associated with microbial dysbiosis, such as the various gastrointestinal disorders and the same colorectal cancer.
Collapse
Affiliation(s)
- Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- SOD of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi (AOUC), 50134 Florence, Italy
| | - Clemente Capasso
- CNR, Institute of Biosciences and Bioresources, 80131 Napoli, Italy
| | - Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | | |
Collapse
|
94
|
Mackie GM, Copland A, Takahashi M, Nakanishi Y, Everard I, Kato T, Oda H, Kanaya T, Ohno H, Maslowski KM. Bacterial cancer therapy in autochthonous colorectal cancer affects tumor growth and metabolic landscape. JCI Insight 2021; 6:e139900. [PMID: 34710062 PMCID: PMC8675204 DOI: 10.1172/jci.insight.139900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 10/27/2021] [Indexed: 12/21/2022] Open
Abstract
Bacterial cancer therapy (BCT) shows great promise for treatment of solid tumors, yet basic mechanisms of bacterial-induced tumor suppression remain undefined. Attenuated strains of Salmonella enterica serovar Typhimurium (STm) have commonly been used in mouse models of BCT in xenograft and orthotopic transplant cancer models. We aimed to better understand the tumor epithelium-targeted mechanisms of BCT by using autochthonous mouse models of intestinal cancer and tumor organoid cultures to assess the effectiveness and consequences of oral treatment with aromatase A-deficient STm (STmΔaroA). STmΔaroA delivered by oral gavage significantly reduced tumor burden and tumor load in both a colitis-associated colorectal cancer (CAC) model and in a spontaneous Apcmin/+ intestinal cancer model. STmΔaroA colonization of tumors caused alterations in transcription of mRNAs associated with tumor stemness, epithelial-mesenchymal transition, and cell cycle. Metabolomic analysis of tumors demonstrated alteration in the metabolic environment of STmΔaroA-treated tumors, suggesting that STmΔaroA imposes metabolic competition on the tumor. Use of tumor organoid cultures in vitro recapitulated effects seen on tumor stemness, mesenchymal markers, and altered metabolome. Furthermore, live STmΔaroA was required, demonstrating active mechanisms including metabolite usage. We have demonstrated that oral BCT is efficacious in autochthonous intestinal cancer models, that BCT imposes metabolic competition, and that BCT has direct effects on the tumor epithelium affecting tumor stem cells.
Collapse
Affiliation(s)
- Gillian M. Mackie
- University of Birmingham, Institute of Immunology and Immunotherapy and Institute of Metabolism and Systems Research, Birmingham, United Kingdom
| | - Alastair Copland
- University of Birmingham, Institute of Immunology and Immunotherapy and Institute of Metabolism and Systems Research, Birmingham, United Kingdom
| | - Masumi Takahashi
- Laboratory for Intestinal Ecosystem, RIKEN Institute for Integrative Medical Science, Yokohama, Japan
| | - Yumiko Nakanishi
- Laboratory for Intestinal Ecosystem, RIKEN Institute for Integrative Medical Science, Yokohama, Japan
- Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Isabel Everard
- University of Birmingham, Institute of Immunology and Immunotherapy and Institute of Metabolism and Systems Research, Birmingham, United Kingdom
| | - Tamotsu Kato
- Laboratory for Intestinal Ecosystem, RIKEN Institute for Integrative Medical Science, Yokohama, Japan
- Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Hirotsugu Oda
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Inflammatory Disease Section, National Human Genome Research Institute, NIH, Bethesda, Maryland, USA
| | - Takashi Kanaya
- Laboratory for Intestinal Ecosystem, RIKEN Institute for Integrative Medical Science, Yokohama, Japan
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Institute for Integrative Medical Science, Yokohama, Japan
- Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Kendle M. Maslowski
- University of Birmingham, Institute of Immunology and Immunotherapy and Institute of Metabolism and Systems Research, Birmingham, United Kingdom
- Laboratory for Intestinal Ecosystem, RIKEN Institute for Integrative Medical Science, Yokohama, Japan
| |
Collapse
|
95
|
Efficacy of first-line atezolizumab combination therapy in patients with non-small cell lung cancer receiving proton pump inhibitors: post hoc analysis of IMpower150. Br J Cancer 2021; 126:42-47. [PMID: 34711947 DOI: 10.1038/s41416-021-01606-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 10/07/2021] [Accepted: 10/19/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Proton pump inhibitors (PPIs) are commonly used concomitant to cancer treatment and they induce gut microbiota changes. It is increasingly apparent that gut dysbiosis can reduce the effectiveness of immune checkpoint inhibitors (ICI). However, little is known about PPI effects on outcomes with ICIs, particularly in combination, ICI approaches. METHODS Post hoc, Cox proportional hazard analysis of phase III trial, IMpower150 was conducted to assess the association between PPI use and overall survival (OS) and progression-free survival (PFS) in chemotherapy-naive, metastatic non-squamous non-small cell lung cancer participants randomised atezolizumab plus carboplatin plus paclitaxel (ACP), bevacizumab plus carboplatin plus paclitaxel (BCP), or atezolizumab plus BCP (ABCP). PPI use was defined as any PPI administration between 30 days prior and 30 days after treatment initiation. RESULTS Of 1202 participants, 441 (37%) received a PPI. PPI use was independently associated with worse OS (n = 748; hazard ratio (HR) [95% confidence interval (CI)] = 1.53 [1.21-1.95], P < 0.001) and PFS (1.34 [1.12-1.61], P = 0.002) in the pooled atezolizumab arms (ACP plus ABCP). This association was not apparent for BCP (n = 368; OS 1.01 [0.73-1.39], P = 0.969; PFS 0.97 [0.76-1.25], P = 0.827). The observed OS treatment effect (HR 95% CI) of the atezolizumab (ACP plus ABCP) arms vs BCP was 1.03 (0.77-1.36) for PPI users compared to 0.68 (0.54-0.86) for non-users (P [interaction] = 0.028). A similar association was noted for ABCP vs BCP (PPI users 0.96 [0.68-1.35]; PPI non-users 0.66 [0.50-0.87]; P [interaction] = 0.095). CONCLUSIONS PPI use was a negative prognostic marker in patients treated with ACP or ABCP, but not BCP. The analysis suggests that PPIs negatively influence the magnitude of ICI efficacy.
Collapse
|
96
|
Rapoport BL, Shannon VR, Cooksley T, Johnson DB, Anderson L, Blidner AG, Tintinger GR, Anderson R. Pulmonary Toxicities Associated With the Use of Immune Checkpoint Inhibitors: An Update From the Immuno-Oncology Subgroup of the Neutropenia, Infection & Myelosuppression Study Group of the Multinational Association for Supportive Care in Cancer. Front Pharmacol 2021; 12:743582. [PMID: 34675810 PMCID: PMC8523897 DOI: 10.3389/fphar.2021.743582] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 09/20/2021] [Indexed: 12/19/2022] Open
Abstract
The development of immune checkpoint inhibitors (ICIs) has revolutionized cancer treatment, with agents such as nivolumab, pembrolizumab, and cemiplimab targeting programmed cell death protein-1 (PD-1) and durvalumab, avelumab, and atezolizumab targeting PD-ligand 1 (PD-L1). Ipilimumab targets cytotoxic T lymphocyte-associated antigen-4 (CTLA-4). These inhibitors have shown remarkable efficacy in melanoma, lung cancer, urothelial cancer, and a variety of solid tumors, either as single agents or in combination with other anticancer modalities. Additional indications are continuing to evolve. Checkpoint inhibitors are associated with less toxicity when compared to chemotherapy. These agents enhance the antitumor immune response and produce side- effects known as immune-related adverse events (irAEs). Although the incidence of immune checkpoint inhibitor pneumonitis (ICI-Pneumonitis) is relatively low, this complication is likely to cause the delay or cessation of immunotherapy and, in severe cases, may be associated with treatment-related mortality. The primary mechanism of ICI-Pneumonitis remains unclear, but it is believed to be associated with the immune dysregulation caused by ICIs. The development of irAEs may be related to increased T cell activity against cross-antigens expressed in tumor and normal tissues. Treatment with ICIs is associated with an increased number of activated alveolar T cells and reduced activity of the anti-inflammatory Treg phenotype, leading to dysregulation of T cell activity. This review discusses the pathogenesis of alveolar pneumonitis and the incidence, diagnosis, and clinical management of pulmonary toxicity, as well as the pulmonary complications of ICIs, either as monotherapy or in combination with other anticancer modalities, such as thoracic radiotherapy.
Collapse
Affiliation(s)
- Bernardo L Rapoport
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.,The Medical Oncology Centre of Rosebank, Johannesburg, South Africa.,The Multinational Association for Supportive Care in Cancer (MASCC), Immuno-Oncology Subgroup of the Neutropenia, Infection and Myelosuppression Study Group, Manchester, United Kingdom
| | - Vickie R Shannon
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Tim Cooksley
- The Multinational Association for Supportive Care in Cancer (MASCC), Immuno-Oncology Subgroup of the Neutropenia, Infection and Myelosuppression Study Group, Manchester, United Kingdom.,Manchester University Foundation Trust, Manchester, United Kingdom.,The Christie, University of Manchester, Manchester, United Kingdom
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Centre and Vanderbilt Ingram Cancer Center, Nashville, TN, United States
| | - Lindsay Anderson
- Department of Radiation Oncology, Steve Biko Academic Hospital, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Ada G Blidner
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine, CONICET, Buenos Aires, Argentina
| | - Gregory R Tintinger
- Department of Internal Medicine, Steve Biko Academic Hospital, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.,The Multinational Association for Supportive Care in Cancer (MASCC), Immuno-Oncology Subgroup of the Neutropenia, Infection and Myelosuppression Study Group, Manchester, United Kingdom
| |
Collapse
|
97
|
Cullin N, Azevedo Antunes C, Straussman R, Stein-Thoeringer CK, Elinav E. Microbiome and cancer. Cancer Cell 2021; 39:1317-1341. [PMID: 34506740 DOI: 10.1016/j.ccell.2021.08.006] [Citation(s) in RCA: 243] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/05/2021] [Accepted: 08/13/2021] [Indexed: 12/14/2022]
Abstract
The human microbiome constitutes a complex multikingdom community that symbiotically interacts with the host across multiple body sites. Host-microbiome interactions impact multiple physiological processes and a variety of multifactorial disease conditions. In the past decade, microbiome communities have been suggested to influence the development, progression, metastasis formation, and treatment response of multiple cancer types. While causal evidence of microbial impacts on cancer biology is only beginning to be unraveled, enhanced molecular understanding of such cancer-modulating interactions and impacts on cancer treatment are considered of major scientific importance and clinical relevance. In this review, we describe the molecular pathogenic mechanisms shared throughout microbial niches that contribute to the initiation and progression of cancer. We highlight advances, limitations, challenges, and prospects in understanding how the microbiome may causally impact cancer and its treatment responsiveness, and how microorganisms or their secreted bioactive metabolites may be potentially harnessed and targeted as precision cancer therapeutics.
Collapse
Affiliation(s)
- Nyssa Cullin
- Microbiome and Cancer Division, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Camila Azevedo Antunes
- Microbiome and Cancer Division, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Ravid Straussman
- Department of Molecular Cell Biology, Weizmann Institute of Science, 234 Herzl Street, 7610001 Rehovot, Israel
| | - Christoph K Stein-Thoeringer
- Microbiome and Cancer Division, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | - Eran Elinav
- Microbiome and Cancer Division, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Immunology, Weizmann Institute of Science, 234 Herzl Street, 7610001 Rehovot, Israel.
| |
Collapse
|
98
|
Lee KA, Luong MK, Shaw H, Nathan P, Bataille V, Spector TD. The gut microbiome: what the oncologist ought to know. Br J Cancer 2021; 125:1197-1209. [PMID: 34262150 PMCID: PMC8548300 DOI: 10.1038/s41416-021-01467-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 06/10/2021] [Indexed: 02/06/2023] Open
Abstract
The gut microbiome (GM) has been implicated in a vast number of human pathologies and has become a focus of oncology research over the past 5 years. The normal gut microbiota imparts specific function in host nutrient metabolism, xenobiotic and drug metabolism, maintenance of structural integrity of the gut mucosal barrier, immunomodulation and protection against pathogens. Strong evidence is emerging to support the effects of the GM on the development of some malignancies but also on responses to cancer therapies, most notably, immune checkpoint inhibition. Tools for manipulating the GM including dietary modification, probiotics and faecal microbiota transfer (FMT) are in development. Current understandings of the many complex interrelationships between the GM, cancer, the immune system, nutrition and medication are ultimately based on a combination of short-term clinical trials and observational studies, paired with an ever-evolving understanding of cancer biology. The next generation of personalised cancer therapies focusses on molecular and phenotypic heterogeneity, tumour evolution and immune status; it is distinctly possible that the GM will become an increasingly central focus amongst them. The aim of this review is to provide clinicians with an overview of microbiome science and our current understanding of the role the GM plays in cancer.
Collapse
Affiliation(s)
- K A Lee
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK.
- Department of Medical Oncology, Mount Vernon Hospital, Northwood, UK.
- Department of Medical Oncology, The Royal Marsden, London, UK.
| | - M K Luong
- Department of Medical Oncology, Guy's & St Thomas Hospital, London, UK
| | - H Shaw
- Department of Medical Oncology, Mount Vernon Hospital, Northwood, UK
- Early Phase Trial Unit, Department of Medical Oncology, University College London Hospital, London, UK
| | - P Nathan
- Department of Medical Oncology, Mount Vernon Hospital, Northwood, UK
| | - V Bataille
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- Department of Dermatology, Mount Vernon Hospital, Northwood, UK
| | - T D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| |
Collapse
|
99
|
Terrisse S, Derosa L, Iebba V, Ghiringhelli F, Vaz-Luis I, Kroemer G, Fidelle M, Christodoulidis S, Segata N, Thomas AM, Martin AL, Sirven A, Everhard S, Aprahamian F, Nirmalathasan N, Aarnoutse R, Smidt M, Ziemons J, Caldas C, Loibl S, Denkert C, Durand S, Iglesias C, Pietrantonio F, Routy B, André F, Pasolli E, Delaloge S, Zitvogel L. Intestinal microbiota influences clinical outcome and side effects of early breast cancer treatment. Cell Death Differ 2021; 28:2778-2796. [PMID: 33963313 PMCID: PMC8408230 DOI: 10.1038/s41418-021-00784-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/05/2021] [Accepted: 04/12/2021] [Indexed: 02/03/2023] Open
Abstract
The prognosis of early breast cancer (BC) relies on cell autonomous and immune parameters. The impact of the intestinal microbiome on clinical outcome has not yet been evaluated. Shotgun metagenomics was used to determine the composition of the fecal microbiota in 121 specimens from 76 early BC patients, 45 of whom were paired before and after chemotherapy. These patients were enrolled in the CANTO prospective study designed to record the side effects associated with the clinical management of BC. We analyzed associations between baseline or post-chemotherapy fecal microbiota and plasma metabolomics with BC prognosis, as well as with therapy-induced side effects. We examined the clinical relevance of these findings in immunocompetent mice colonized with BC patient microbiota that were subsequently challenged with histo-compatible mouse BC and chemotherapy. We conclude that specific gut commensals that are overabundant in BC patients compared with healthy individuals negatively impact BC prognosis, are modulated by chemotherapy, and may influence weight gain and neurological side effects of BC therapies. These findings obtained in adjuvant and neoadjuvant settings warrant prospective validation.
Collapse
Affiliation(s)
- Safae Terrisse
- Gustave Roussy Cancer Center, Villejuif, France
- INSERM U1015, Equipe Labellisée par la ligue Contre le Cancer, Villejuif, France
- University Paris Saclay, School of Medicine, Le Kremlin-Bicêtre, France
- Department of Medical Oncology, Saint Louis Hospital, Paris, France
| | - Lisa Derosa
- Gustave Roussy Cancer Center, Villejuif, France
- INSERM U1015, Equipe Labellisée par la ligue Contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France
| | - Valerio Iebba
- Gustave Roussy Cancer Center, Villejuif, France
- INSERM U1015, Equipe Labellisée par la ligue Contre le Cancer, Villejuif, France
| | - François Ghiringhelli
- Research Platform in Biological Oncology, Dijon, France
- GIMI Genetic and Immunology Medical Institute, Dijon, France
- University of Burgundy-Franche Comté, Dijon, France
- Department of Medical Oncology, Center GF Leclerc, Dijon, France
| | - Ines Vaz-Luis
- INSERM U 981, Gustave Roussy, Villejuif, Île-de-France, France
- Department of Medicine, Breast Cancer committee, Gustave Roussy, Villejuif, France
| | - Guido Kroemer
- INSERM U1138, Equipe Labelisée par la ligue Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Cell Biology and Metabolomics Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Université de Paris, Paris, France
- Sorbonne Université, Paris, France
| | - Marine Fidelle
- Gustave Roussy Cancer Center, Villejuif, France
- INSERM U1015, Equipe Labellisée par la ligue Contre le Cancer, Villejuif, France
- University Paris Saclay, School of Medicine, Le Kremlin-Bicêtre, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France
| | - Stergios Christodoulidis
- University Paris Saclay, Saint-Aubain, France
- Prism Precision Medicine Center, Gustave Roussy, Villejuif, France
| | - Nicola Segata
- Department of Medical Sciences, University of Trieste, Trieste, Italy
- Department CIBIO, University of Trento, Trento, Italy
| | | | | | | | | | - Fanny Aprahamian
- Cell Biology and Metabolomics Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | | | - Romy Aarnoutse
- Faculty of Health, Medicine & Life Sciences, Department of Surgery, Maastricht, The Netherlands
- GROW School for Oncology & Developmental Biology, Maastricht, The Netherlands
- Maastricht University, Maastricht, The Netherlands
| | - Marjolein Smidt
- Faculty of Health, Medicine & Life Sciences, Department of Surgery, Maastricht, The Netherlands
- GROW School for Oncology & Developmental Biology, Maastricht, The Netherlands
- Maastricht University, Maastricht, The Netherlands
| | - Janine Ziemons
- Faculty of Health, Medicine & Life Sciences, Department of Surgery, Maastricht, The Netherlands
- GROW School for Oncology & Developmental Biology, Maastricht, The Netherlands
- Maastricht University, Maastricht, The Netherlands
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, UK
| | - Sibylle Loibl
- Goethe University Frankfurt, Frankfurt, Germany
- Clinical Consultant Centre for Haematology and Oncology, Frankfurt, Germany
| | - Carsten Denkert
- Philipps-University Marburg and University Hospital Marburg (UKGM), Marburg, Germany
| | - Sylvere Durand
- Cell Biology and Metabolomics Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Claudia Iglesias
- Cell Biology and Metabolomics Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | | | - Bertrand Routy
- Division d'hémato-oncologie, Département de Médicine, Centre Hospitalier de l'université de Montréal (CHUM), Montréal, Québec, Canada
| | - Fabrice André
- Gustave Roussy Cancer Center, Villejuif, France
- INSERM U 981, Gustave Roussy, Villejuif, Île-de-France, France
- Department of Medicine, Breast Cancer committee, Gustave Roussy, Villejuif, France
- University Paris Saclay, Saint-Aubain, France
| | - Edoardo Pasolli
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Suzette Delaloge
- Gustave Roussy Cancer Center, Villejuif, France
- Department of Medicine, Breast Cancer committee, Gustave Roussy, Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Center, Villejuif, France.
- INSERM U1015, Equipe Labellisée par la ligue Contre le Cancer, Villejuif, France.
- University Paris Saclay, School of Medicine, Le Kremlin-Bicêtre, France.
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France.
| |
Collapse
|
100
|
Acharya M, Kim T, Li C. Broad-Spectrum Antibiotic Use and Disease Progression in Early-Stage Melanoma Patients: A Retrospective Cohort Study. Cancers (Basel) 2021; 13:4367. [PMID: 34503177 PMCID: PMC8431240 DOI: 10.3390/cancers13174367] [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: 08/16/2021] [Accepted: 08/24/2021] [Indexed: 11/16/2022] Open
Abstract
Animal studies and a few clinical studies have reported mixed findings on the association between antibiotics and cancer incidence. Antibiotics may inhibit tumor cell growth, but could also alter the gut-microbiome-modulated immune system and increase the risk of cancer. Studies that assess how antibiotics affect the progression of cancer are limited. We evaluated the association between broad-spectrum antibiotic use and melanoma progression. We conducted a retrospective cohort study using IQVIA PharMetrics® Plus data (2008-2018). We identified patients with malignant melanoma who underwent wide local excision or Mohs micrographic surgery within 90 days of first diagnosis. Surgery date was the index date. Patients were excluded if they had any other cancer diagnosis or autoimmune disorders in 1 year before the index date ("baseline"). Exposure to broad-spectrum antibiotics was identified in three time windows using three cohorts: 3 months prior to the index date, 1 month after the index date, and 3 months after the index date. The covariates were patients' demographic and clinical characteristics identified in the 1-year baseline period. The patients were followed from the index date until cancer progression, loss of enrollment, or the end of 2 years after the index date. Progression was defined as: (i) any hospice care after surgery, (ii) a new round of treatment for melanoma (surgery, chemotherapy, immunotherapy, targeted therapy, or radiotherapy) 180 days after prior treatment, or (iii) a metastasis diagnosis or a diagnosis of a new nonmelanoma primary cancer at least 180 days after first melanoma diagnosis or prior treatment. A high-dimensional propensity score approach with inverse weighting was used to adjust for the patients' baseline differences. Cox proportional hazard regression was used for estimating the association. The final samples included 3930, 3831, and 3587 patients (mean age: 56 years). Exposure to antibiotics was 16% in the prior-3-months, 22% in the post-1-month, and 22% in the post-3-months. In the pre-3-months analysis, 9% of the exposed group and 9% of the unexposed group had progressed. Antibiotic use was not associated with melanoma progression (HR: 0.81; 95% CI: 0.57-1.14). However, antibiotic use in subsequent 1 month and subsequent 3 months was associated with 31% reduction (HR: 0.69; 95% CI: 0.51-0.92) and 32% reduction (HR: 0.68; 95% CI: 0.51-0.91) in progression, respectively. In this cohort of patients with likely early-stage melanoma cancer, antibiotic use in 1 month and 3 months after melanoma surgery was associated with a lower risk of melanoma progression. Future studies are warranted to validate the findings.
Collapse
Affiliation(s)
- Mahip Acharya
- Division of Pharmaceutical Evaluation and Policy, University of Arkansas for Medical Sciences College of Pharmacy, Little Rock, AR 72205, USA;
| | - Thomas Kim
- Department of Radiation Oncology, Rush University Medical College, Chicago, IL 60612, USA;
| | - Chenghui Li
- Division of Pharmaceutical Evaluation and Policy, University of Arkansas for Medical Sciences College of Pharmacy, Little Rock, AR 72205, USA;
| |
Collapse
|