101
|
Spyrou N, Vallianou N, Kadillari J, Dalamaga M. The interplay of obesity, gut microbiome and diet in the immune check point inhibitors therapy era. Semin Cancer Biol 2021; 73:356-376. [DOI: 10.1016/j.semcancer.2021.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 03/22/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022]
|
102
|
Cannarile MA, Gomes B, Canamero M, Reis B, Byrd A, Charo J, Yadav M, Karanikas V. Biomarker Technologies to Support Early Clinical Immuno-oncology Development: Advances and Interpretation. Clin Cancer Res 2021; 27:4147-4159. [PMID: 33766813 DOI: 10.1158/1078-0432.ccr-20-2345] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/02/2021] [Accepted: 03/08/2021] [Indexed: 11/16/2022]
Abstract
Today, there is a huge effort to develop cancer immunotherapeutics capable of combating cancer cells as well as the biological environment in which they can grow, adapt, and survive. For such treatments to benefit more patients, there is a great need to dissect the complex interplays between tumor cells and the host's immune system. Monitoring mechanisms of resistance to immunotherapeutics can delineate the evolution of key players capable of driving an efficacious antitumor immune response. In doing so, simultaneous and systematic interrogation of multiple biomarkers beyond single biomarker approaches needs to be undertaken. Zooming into cell-to-cell interactions using technological advancements with unprecedented cellular resolution such as single-cell spatial transcriptomics, advanced tissue histology approaches, and new molecular immune profiling tools promises to provide a unique level of molecular granularity of the tumor environment and may support better decision-making during drug development. This review will focus on how such technological tools are applied in clinical settings, to inform the underlying tumor-immune biology of patients and offer a deeper understanding of cancer immune responsiveness to immuno-oncology treatments.
Collapse
Affiliation(s)
- Michael A Cannarile
- F. Hoffmann-La Roche AG, Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Munich, Munich, Germany
| | - Bruno Gomes
- F. Hoffmann-La Roche AG, Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Basel, Basel, Switzerland
| | - Marta Canamero
- F. Hoffmann-La Roche AG, Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Munich, Munich, Germany
| | - Bernhard Reis
- F. Hoffmann-La Roche AG, Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Basel, Basel, Switzerland
| | | | - Jehad Charo
- F. Hoffmann-La Roche AG, Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Zurich, Zurich, Switzerland
| | | | - Vaios Karanikas
- F. Hoffmann-La Roche AG, Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Zurich, Zurich, Switzerland.
| |
Collapse
|
103
|
Clinical prediction of bacteremia and early antibiotics therapy in patients with solid tumors. Infect Control Hosp Epidemiol 2021; 43:1112-1118. [PMID: 34315562 DOI: 10.1017/ice.2021.314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To investigate the relationship between the systemic inflammatory response syndrome (SIRS), early antibiotic use, and bacteremia in solid-tumor patients. DESIGN, SETTING, AND PARTICIPANTS We conducted a retrospective observational study of adults with solid tumors admitted to a tertiary-care hospital through the emergency department over a 2-year period. Patients with neutropenic fever, organ transplant, trauma, or cardiopulmonary arrest were excluded. METHODS Rates of SIRS, bacteremia, and early antibiotics (initiation within 8 hours of presentation) were compared using the χ2 and Student t tests. Binomial regression and receiver operator curves were analyzed to assess predictors of bacteremia and early antibiotics. RESULTS Early antibiotics were administered in 507 (37%) of 1,344 SIRS-positive cases and 492 (22%) of 2,236 SIRS-negative cases (P < .0001). Of SIRS-positive cases, 70% had blood cultures drawn within 48 hours and 19% were positive; among SIRS negative cases, 35% had cultures and 13% were positive (19% vs 13%; P = .003). Bacteremic cases were more often SIRS positive than nonbacteremic cases (60% vs 50%; P =.003), but they received early antibiotics at similar rates (50% vs 49%, P = .72). Three SIRS components predicted early antibiotics: temperature (OR, 1.7; 95% CI, 1.31-2.29; P = .0001), tachycardia (OR, 1.4; 95% CI, 1.10-1.69; P < .0001), and white blood-cell count (OR, 1.8; 95% CI, 1.56-2.14; P < .0001). Only temperature (OR, 1.6; 95% CI, 1.09-2.41; P = .01) and tachycardia (OR, 1.5; 95% CI, 1.09-2.06; P = .01) predicted bacteremia. SIRS criteria as a composite were poorly predictive of bacteremia (AUC, 0.57). CONCLUSIONS SIRS criteria are frequently used to determine the need for early antibiotics, but they are poor predictors of bacteremia in solid-tumor patients. More reliable models are needed to guide judicious use of antibiotics in this population.
Collapse
|
104
|
Mohindroo C, Hasanov M, Rogers JE, Dong W, Prakash LR, Baydogan S, Mizrahi JD, Overman MJ, Varadhachary GR, Wolff RA, Javle MM, Fogelman DR, Lotze MT, Kim MP, Katz MHG, Pant S, Tzeng CWD, McAllister F. Antibiotic use influences outcomes in advanced pancreatic adenocarcinoma patients. Cancer Med 2021; 10:5041-5050. [PMID: 34250759 PMCID: PMC8335807 DOI: 10.1002/cam4.3870] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/14/2020] [Accepted: 12/31/2020] [Indexed: 12/21/2022] Open
Abstract
Recent studies defined a potentially important role of the microbiome in modulating pancreatic ductal adenocarcinoma (PDAC) and responses to therapies. We hypothesized that antibiotic usage may predict outcomes in patients with PDAC. We retrospectively analyzed clinical data of patients with resectable or metastatic PDAC seen at MD Anderson Cancer from 2003 to 2017. Demographic, chemotherapy regimen and antibiotic use, duration, type, and reason for indication were recorded. A total of 580 patients with PDAC were studied, 342 resected and 238 metastatic patients, selected retrospectively from our database. Antibiotic use, for longer than 48 hrs, was detected in 209 resected patients (61%) and 195 metastatic ones (62%). On resectable patients, we did not find differences in overall survival (OS) or progression‐free survival (PFS), based on antibiotic intake. However, in the metastatic cohort, antibiotic consumption was associated with a significantly longer OS (13.3 months vs. 9.0 months, HR 0.48, 95% CI 0.34–0.7, p = 0.0001) and PFS (4.4 months vs. 2 months, HR 0.48, 95% CI 0.34–0.68, p = <0.0001). In multivariate analysis, the impact of ATB remained significant for PFS (HR 0.59, p = 0.005) and borderline statistically significant for OS (HR 0.69, p = 0.06). When we analyzed by chemotherapy regimen, we found that patients who received gemcitabine‐based chemotherapy as first‐line therapy (n = 118) had significantly prolonged OS (HR 0.4, p 0.0013) and PFS (HR 0.55, p 0.02) if they received antibiotics, while those receiving 5FU‐based chemotherapy (n = 98) had only prolonged PFS (HR 0.54, p = 0.03). Antibiotics‐associated modulation of the microbiome is associated with better outcomes in patients with metastatic PDAC. We have analyzed the effect of antibiotics’ intake on two cohorts of patients with pancreatic adenocarcinoma, resectable, and metastatic. We have found that on the metastatic cohort, antibiotics use was significantly associated with better outcomes, particularly, on patients that received gemcitabine based‐chemotherapy as the first line.
Collapse
Affiliation(s)
- Chirayu Mohindroo
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Merve Hasanov
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jane E Rogers
- Pharmacy Clinical Programs, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wenli Dong
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Laura R Prakash
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Seyda Baydogan
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jonathan D Mizrahi
- Department of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael J Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gauri R Varadhachary
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Robert A Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Milind M Javle
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David R Fogelman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Michael P Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Matthew H G Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shubham Pant
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Investigation Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ching-Wei D Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Florencia McAllister
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
105
|
Malczewski AB, Ketheesan N, Coward JIG, Navarro S. Enhancing Checkpoint Inhibitor Therapy in Solid Tissue Cancers: The Role of Diet, the Microbiome & Microbiome-Derived Metabolites. Front Immunol 2021; 12:624434. [PMID: 34305883 PMCID: PMC8293987 DOI: 10.3389/fimmu.2021.624434] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 06/24/2021] [Indexed: 12/14/2022] Open
Abstract
Host immunity plays a central role in the regulation of anti-tumour responses during checkpoint inhibitor therapy (CIT). The mechanisms involved in long lasting remission remain unclear. Animal studies have revealed that the microbiome influences the host immune response. This is supported by human studies linking a higher microbial richness and diversity with enhanced responses to CIT. This review focuses on the role of diet, the microbiome and the microbiome-derived metabolome in enhancing responses to current CIT in solid tissue cancers. The Western diet has been associated with dysbiosis, inflammation and numerous metabolic disorders. There is preliminary evidence that lifestyle factors including a high fibre diet are associated with improved responses to CIT via a potential effect on the microbiota. The mechanisms through which the microbiota may regulate long-term immunotherapy responses have yet to be determined, although bacterial-metabolites including short chain fatty acids (SCFAs) are recognized to have an impact on T cell differentiation, and may affect T effector/regulatory T cell balance. SCFAs were also shown to enhance the memory potential of activated CD8 T cells. Many therapeutic approaches including dietary manipulation and fecal transplantation are currently being explored in order to enhance immunotherapy responses. The microbiome-derived metabolome may be one means through which bacterial metabolic products can be monitored from the start of treatment and could be used to identify patients at risk of poor immunotherapy responses. The current review will discuss recent advances and bring together literature from related fields in nutrition, oncology and immunology to discuss possible means of modulating immunity to improve responses to current CIT.
Collapse
Affiliation(s)
- Agnieszka Beata Malczewski
- Icon Cancer Centre, Wesley, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Science and Technology, University of New England, Armidale, NSW, Australia
| | - Natkunam Ketheesan
- Science and Technology, University of New England, Armidale, NSW, Australia
| | - Jermaine I. G. Coward
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Icon Cancer Centre, South Brisbane, Brisbane, QLD, Australia
| | - Severine Navarro
- Department of Immunology, QIMR Berghofer, Brisbane, QLD, Australia
- Woolworths Centre for Childhood Nutrition Research, Faculty of Health, Queensland University of Technology, South Brisbane, QLD, Australia
| |
Collapse
|
106
|
Influence of immunomodulatory drugs on the gut microbiota. Transl Res 2021; 233:144-161. [PMID: 33515779 PMCID: PMC8184576 DOI: 10.1016/j.trsl.2021.01.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/10/2020] [Accepted: 01/26/2021] [Indexed: 12/17/2022]
Abstract
Immunomodulatory medications are a mainstay of treatment for autoimmune diseases and malignancies. In addition to their direct effects on immune cells, these medications also impact the gut microbiota. Drug-induced shifts in commensal microbes can lead to indirect but important changes in the immune response. We performed a comprehensive literature search focusing on immunotherapy/microbe interactions. Immunotherapies were categorized into 5 subtypes based on their mechanisms of action: cell trafficking inhibitors, immune checkpoint inhibitors, immunomodulators, antiproliferative drugs, and inflammatory cytokine inhibitors. Although no consistent relationships were observed between types of immunotherapy and microbiota, most immunotherapies were associated with shifts in specific colonizing bacterial taxa. The relationships between colonizing microbes and drug efficacy were not well-studied for autoimmune diseases. In contrast, the efficacy of immune checkpoint inhibitors for cancer was tied to the baseline composition of the gut microbiota. There was a paucity of high-quality data; existing data were generated using heterogeneous sampling and analytic techniques, and most studies involved small numbers of participants. Further work is needed to elucidate the extent and clinical significance of immunotherapy effects on the human microbiome.
Collapse
|
107
|
Impact of Use of Antibiotics on Response to Immune Checkpoint Inhibitors and Tumor Microenvironment. Am J Clin Oncol 2021; 44:247-253. [PMID: 33826550 DOI: 10.1097/coc.0000000000000813] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Antibiotic use can result in reduced efficacy of immune checkpoint blockade (ICB), presumably because of dysbiosis of the intestinal microbiome. We sought to determine the precise temporal relation between antibiotic therapy and its possible effects on ICB efficacy. We also investigated the histologic changes in the tumor microenvironment secondary to antibiotics use. METHODS AND OBJECTIVES This was a single institution retrospective study that evaluated the impact of antibiotics on outcomes of patients with advanced or metastatic malignancy who were treated with ICB. Use of antibiotics among patients treated with ICB was assessed during a 12-week period before and after initiation of ICB. The primary outcome was response to ICB. Histologic changes in the tumor microenvironment following antibiotics use were also examined. RESULTS Between January 1, 2011 and December 31, 2018, 414 patients were identified who received ICB, and 207 patients (50%) received antibiotics within 12 weeks (before/after) of initiation of ICB. In univariate analysis, antibiotic use following initiation of ICB was associated with a significantly reduced response (odds ratio [OR]: 0.33, 95% confidence interval [CI]: 0.2-0.52, P<0.001). There was no significant negative impact on response to immunotherapy when antibiotics were used before ICB initiation (OR: 0.87, 95% CI: 0.55-1.34, P=0.52). The maximal negative impact of antibiotics occurred in the first 6 weeks after initiating ICB, and was independently associated with significantly reduced likelihood of response to immunotherapy in multivariable analysis (OR: 0.48, 95% CI: 0.29-0.8, P=0.01). CONCLUSIONS This study demonstrates that the use of antibiotics during ICB significantly negatively impacts the efficacy of immunotherapy. The maximal negative impact occurs if the antibiotics are used in the first 6 weeks after initiating ICB.
Collapse
|
108
|
Shi Z, Zhang M. Emerging Roles for the Gut Microbiome in Lymphoid Neoplasms. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2021; 15:11795549211024197. [PMID: 34211309 PMCID: PMC8216388 DOI: 10.1177/11795549211024197] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 05/18/2021] [Indexed: 12/15/2022]
Abstract
Lymphoid neoplasms encompass a heterogeneous group of malignancies with a predilection for immunocompromised individuals, and the disease burden of lymphoid neoplasms has been rising globally over the last decade. At the same time, mounting studies delineated a crucial role of the gut microbiome in the aetiopathogenesis of various diseases. Orchestrated interactions between myriad microorganisms and the gastrointestinal mucosa establish a defensive barrier for a range of physiological processes, especially immunity and metabolism. These findings provide new perspectives to harness our knowledge of the gut microbiota for preclinical and clinical studies of lymphoma. Here, we review recent findings that support a role for the gut microbiota in the development of lymphoid neoplasms and pinpoint relevant molecular mechanisms. Accordingly, we propose the microbiota-gut-lymphoma axis as a promising target for clinical translation, including auxiliary diagnosis, novel prevention and treatment strategies, and predicting clinical outcomes and treatment-related adverse effects of the disease in the future. This review will reveal a fascinating avenue of research in the microbiota-mediated lymphoma field.
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
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, China
| |
Collapse
|
109
|
Antibiotic-induced disruption of the microbiome exacerbates chemotherapy-induced diarrhoea and can be mitigated with autologous faecal microbiota transplantation. Eur J Cancer 2021; 153:27-39. [PMID: 34130227 DOI: 10.1016/j.ejca.2021.05.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Chemotherapy is well documented to disrupt the gut microbiome, leading to poor treatment outcomes and a heightened risk of adverse toxicity. Although strong associations exist between its composition and gastrointestinal toxicity, its causal contribution remains unclear. Our inability to move beyond association has limited the development and implementation of microbial-based therapeutics in chemotherapy adjuncts with no clear rationale of how and when to deliver them. METHODS/RESULTS Here, we investigate the impact of augmenting the gut microbiome on gastrointestinal toxicity caused by the chemotherapeutic agent, methotrexate (MTX). Faecal microbiome transplantation (FMT) delivered after MTX had no appreciable impact on gastrointestinal toxicity. In contrast, disruption of the microbiome with antibiotics administered before chemotherapy exacerbated gastrointestinal toxicity, impairing mucosal recovery (P < 0.0001) whilst increasing diarrhoea severity (P = 0.0007) and treatment-related mortality (P = 0.0045). Importantly, these detrimental effects were reversed when the microbiome was restored using autologous FMT (P = 0.03), a phenomenon dictated by the uptake and subsequent expansion of Muribaculaceae. CONCLUSIONS These are the first data to show that clinically impactful symptoms of gastrointestinal toxicity are dictated by the microbiome and provide a clear rationale for how and when to target the microbiome to mitigate the acute and chronic complications caused by disruption of the gastrointestinal microenvironment. Translation of this new knowledge should focus on stabilising and strengthening the gut microbiome before chemotherapy and developing new microbial approaches to accelerate recovery of the mucosa. By controlling the depth and duration of mucosal injury, secondary consequences of gastrointestinal toxicity may be avoided.
Collapse
|
110
|
Antibiotics and steroids, the double enemies of anticancer immunotherapy: a review of the literature. Cancer Immunol Immunother 2021; 70:1511-1517. [PMID: 33165628 DOI: 10.1007/s00262-020-02786-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023]
Abstract
The advent of immunotherapy in onco-haematology has represented a kind of revolution that has been able to modify the prognosis of numerous tumours that until recently would have been rapidly lethal. While much is known about the mechanism of action of these drugs, relatively little is known about the factors that represent potential predictors of response and toxicity. Among these factors, the simultaneous administration of antibiotics and/or steroids seems to have a negative impact. Furthermore, several retrospective studies have highlighted the strong link between cancer and gut microbiota, regardless of the tumour site, and how microbiota, playing a key role in the prevention of systemic inflammation at various levels and in the intestinal homeostasis, can be negatively influenced by the dysbiosis caused by antibiotic therapy administered during or in the weeks immediately preceding the start of immunotherapy. Moreover, we assume that the concurrent administration of steroids, which is often necessary in cancer patients, likely results in a deleterious effect on the therapeutic outcomes of immunotherapy. In this review, we will try to clarify the evidence on the possible detrimental effects of antibiotics and steroids, which are currently considered the double enemies of anticancer immunotherapy.
Collapse
|
111
|
Schubert ML, Rohrbach R, Schmitt M, Stein-Thoeringer CK. The Potential Role of the Intestinal Micromilieu and Individual Microbes in the Immunobiology of Chimeric Antigen Receptor T-Cell Therapy. Front Immunol 2021; 12:670286. [PMID: 34135898 PMCID: PMC8200823 DOI: 10.3389/fimmu.2021.670286] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/04/2021] [Indexed: 12/25/2022] Open
Abstract
Cellular immunotherapy with chimeric antigen receptor (CAR)-T cells (CARTs) represents a breakthrough in the treatment of hematologic malignancies. CARTs are genetically engineered hybrid receptors that combine antigen-specificity of monoclonal antibodies with T cell function to direct patient-derived T cells to kill malignant cells expressing the target (tumor) antigen. CARTs have been introduced into clinical medicine as CD19-targeted CARTs for refractory and relapsed B cell malignancies. Despite high initial response rates, current CART therapies are limited by a long-term loss of antitumor efficacy, the occurrence of toxicities, and the lack of biomarkers for predicting therapy and toxicity outcomes. In the past decade, the gut microbiome of mammals has been extensively studied and evidence is accumulating that human health, apart from our own genome, largely depends on microbes that are living in and on the human body. The microbiome encompasses more than 1000 bacterial species who collectively encode a metagenome that guides multifaceted, bidirectional host-microbiome interactions, primarily through the action of microbial metabolites. Increasing knowledge has been accumulated on the role of the gut microbiome in T cell-driven anticancer immunotherapy. It has been shown that antibiotics, dietary components and gut microbes reciprocally affect the efficacy and toxicity of allogeneic hematopoietic cell transplantation (allo HCT) as the prototype of T cell-based immunotherapy for hematologic malignancies, and that microbiome diversity metrics can predict clinical outcomes of allo HCTs. In this review, we will provide a comprehensive overview of the principles of CD19-CART immunotherapy and major aspects of the gut microbiome and its modulators that impact antitumor T cell transfer therapies. We will outline i) the extrinsic and intrinsic variables that can contribute to the complex interaction of the gut microbiome and host in CART immunotherapy, including ii) antibiotic administration affecting loss of colonization resistance, expansion of pathobionts and disturbed mucosal and immunological homeostasis, and ii) the role of specific gut commensals and their microbial virulence factors in host immunity and inflammation. Although the role of the gut microbiome in CART immunotherapy has only been marginally explored so far, this review may open a new chapter and views on putative connections and mechanisms.
Collapse
Affiliation(s)
- Maria-Luisa Schubert
- Klinik fuer Haematologie, Onkologie und Rheumatologie, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Roman Rohrbach
- Research Division Microbiome and Cancer, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Michael Schmitt
- Klinik fuer Haematologie, Onkologie und Rheumatologie, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Christoph K Stein-Thoeringer
- Research Division Microbiome and Cancer, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany.,Klinik fuer Medizinische Onkologie, Nationales Centrum für Tumorerkrankungen (NCT), Heidelberg, Germany
| |
Collapse
|
112
|
Geum MJ, Kim C, Kang JE, Choi JH, Kim JS, Son ES, Lim SM, Rhie SJ. Broad-Spectrum Antibiotic Regimen Affects Survival in Patients Receiving Nivolumab for Non-Small Cell Lung Cancer. Pharmaceuticals (Basel) 2021; 14:ph14050445. [PMID: 34066877 PMCID: PMC8151442 DOI: 10.3390/ph14050445] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 01/17/2023] Open
Abstract
Antibiotic-induced dysbiosis may affect the efficacy of immune checkpoint inhibitors. We investigated the impact of antibiotics on the clinical outcomes of nivolumab in patients with non-small cell lung cancer (NSCLC). Patients who received nivolumab for NSCLC between July 2015 and June 2018 and who were followed up until June 2020 were included in a retrospective cohort analysis. Of 140 eligible patients, 70 were on antibiotics. Overall survival (OS) was shorter in patients on antibiotics (ABX) compared to those not on antibiotics (NoABX) (p = 0.014). OS was negatively associated with piperacillin/tazobactam (PTZ) (HR = 3.31, 95% CI: 1.77–6.18), days of therapy (DOT) ≥ 2 weeks (HR = 2.56, 95% CI: 1.30–5.22) and DOT of PTZ. The defined daily dose (DDD) in PTZ (r = 0.27) and glycopeptides (r = 0.21) showed weak correlations with mortality. There was no difference in progression-free survival (PFS) between ABX and NoABX; however, PFS was negatively associated with the antibiotic class PTZ and DOT of PTZ. Therefore, the use of a broad-spectrum antibiotic, such as PTZ, the long-term use of antibiotics more than 2 weeks in total and the large amount of defined daily dose of specific antibiotics were associated with decreased survival in patients receiving nivolumab for NSCLC.
Collapse
Affiliation(s)
- Min Jung Geum
- The Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea;
- Department of Pharmacy, Severance Hospital, Yonsei University Health System, Seoul 03722, Korea; (J.S.K.); (E.S.S.)
| | - Chungsoo Kim
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea;
| | - Ji Eun Kang
- College of Pharmacy, Ewha Womans University, Seoul 03760, Korea;
- Department of Pharmacy, National Medical Center, Seoul 04564, Korea
| | - Jae Hee Choi
- Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea;
- Department of Pharmacy, Konkuk University Medical Center, Seoul 05030, Korea
| | - Jae Song Kim
- Department of Pharmacy, Severance Hospital, Yonsei University Health System, Seoul 03722, Korea; (J.S.K.); (E.S.S.)
| | - Eun Sun Son
- Department of Pharmacy, Severance Hospital, Yonsei University Health System, Seoul 03722, Korea; (J.S.K.); (E.S.S.)
| | - Sun Min Lim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Yonsei Cancer Center, Seoul 03722, Korea;
| | - Sandy Jeong Rhie
- The Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea;
- College of Pharmacy, Ewha Womans University, Seoul 03760, Korea;
- Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea;
- Correspondence:
| |
Collapse
|
113
|
Cvetkovic L, Régis C, Richard C, Derosa L, Leblond A, Malo J, Messaoudene M, Desilets A, Belkaid W, Elkrief A, Routy B, Juneau D. Physiologic colonic uptake of 18F-FDG on PET/CT is associated with clinical response and gut microbiome composition in patients with advanced non-small cell lung cancer treated with immune checkpoint inhibitors. Eur J Nucl Med Mol Imaging 2021; 48:1550-1559. [PMID: 33128571 DOI: 10.1007/s00259-020-05081-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) represent the backbone treatment for advanced non-small cell lung cancer (NSCLC). Emerging data suggest that increased gut microbiome diversity is associated with favorable response to ICI and that antibiotic-induced dysbiosis is associated with deleterious outcomes. 18F-FDG physiologic colonic uptake on PET/CT increases following treatment with antibiotics (ATB) and could act as a surrogate marker for microbiome composition and predict prognosis. The aim of this study was to determine if 18F-FDG physiologic colonic uptake prior to ICI initiation correlates with gut microbiome profiling and clinical outcomes in patients with advanced NSCLC. METHODS Seventy-one patients with advanced NSCLC who underwent a PET/CT prior to ICI were identified. Blinded colonic contouring was performed for each colon segment and patients were stratified according to the median of the average colon SUVmax as well as for each segment in low vs. high SUVmax groups. Response rate, progression-free survival (PFS), and overall survival (OS) were compared in the low vs. high SUVmax groups. Gut microbiome composition was analyzed for 23 patients using metagenomics sequencing. RESULTS The high colon SUVmax group had a higher proportion of non-responders (p = 0.033) and significantly shorter PFS (4.1 vs. 11.3 months, HR 1.94, 95% CI 1.11-3.41, p = 0.005). High caecum SUVmax correlated with numerically shorter OS (10.8 vs. 27.6 months, HR 1.85, 95% CI 0.97-3.53, p = 0.058). Metagenomics sequencing revealed distinctive microbiome populations in each group. Patients with low caecum SUVmax had higher microbiome diversity (p = 0.046) and were enriched with Bifidobacteriaceae, Lachnospiraceae, and Bacteroidaceae. CONCLUSIONS Lower colon physiologic 18F-FDG uptake on PET/CT prior to ICI initiation was associated with better clinical outcomes and higher gut microbiome diversity in patients with advanced NSCLC. Here, we propose that 18F-FDG physiologic colonic uptake on PET/CT could serve as a potential novel marker of gut microbiome composition and may predict clinical outcomes in this population.
Collapse
Affiliation(s)
- Lena Cvetkovic
- Department of Hematology and Oncology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, Québec, Canada
| | - Claudine Régis
- Department of Radiology and Nuclear Medicine, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Department of Radiology and Nuclear Medicine, Institut de Cardiologie de Montréal, Montréal, Québec, Canada
| | - Corentin Richard
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, Québec, Canada
| | - Lisa Derosa
- Department of Tumor Immunology and Immunotherapy INSERM U1015, Gustave Roussy Cancer Center, Villejuif, France
| | - Antoine Leblond
- Department of Radiology and Nuclear Medicine, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Department of Radiology and Nuclear Medicine, Centre Hospitalier Affilié Universitaire Régional (CHAUR), Trois-Rivières, Québec, Canada
| | - Julie Malo
- Department of Hematology and Oncology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, Québec, Canada
| | - Meriem Messaoudene
- Department of Hematology and Oncology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, Québec, Canada
| | - Antoine Desilets
- Department of Hematology and Oncology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, Québec, Canada
| | - Wiam Belkaid
- Department of Hematology and Oncology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, Québec, Canada
| | - Arielle Elkrief
- Department of Hematology and Oncology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, Québec, Canada
| | - Bertrand Routy
- Department of Hematology and Oncology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada.
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, Québec, Canada.
| | - Daniel Juneau
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, Québec, Canada.
- Department of Radiology and Nuclear Medicine, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada.
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
114
|
Translational model of melphalan-induced gut toxicity reveals drug-host-microbe interactions that drive tissue injury and fever. Cancer Chemother Pharmacol 2021; 88:173-188. [PMID: 33877390 PMCID: PMC8236460 DOI: 10.1007/s00280-021-04273-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/30/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE Conditioning therapy with high-dose melphalan (HDM) is associated with a high risk of gut toxicity, fever and infections in haematopoietic stem cell transplant (HSCT) recipients. However, validated preclinical models that adequately reflect clinical features of melphalan-induced toxicity are not available. We therefore aimed to develop a novel preclinical model of melphalan-induced toxicity that reflected well-defined clinical dynamics, as well as to identify targetable mechanisms that drive intestinal injury. METHODS Male Wistar rats were treated with 4-8 mg/kg melphalan intravenously. The primary endpoint was plasma citrulline. Secondary endpoints included survival, weight loss, diarrhea, food/water intake, histopathology, body temperature, microbiota composition (16S sequencing) and bacterial translocation. RESULTS Melphalan 5 mg/kg caused self-limiting intestinal injury, severe neutropenia and fever while impairing the microbial metabolome, prompting expansion of enteric pathogens. Intestinal inflammation was characterized by infiltration of polymorphic nuclear cells in the acute phases of mucosal injury, driving derangement of intestinal architecture. Ileal atrophy prevented bile acid reabsorption, exacerbating colonic injury via microbiota-dependent mechanisms. CONCLUSION We developed a novel translational model of melphalan-induced toxicity, which has excellent homology with the well-known clinical features of HDM transplantation. Application of this model will accelerate fundamental and translational study of melphalan-induced toxicity, with the clinical parallels of this model ensuring a greater likelihood of clinical success.
Collapse
|
115
|
Luo B, Zhang Y, Zhang C, Liu X, Shi C. Intestinal microbiota: A potential target for enhancing the antitumor efficacy and reducing the toxicity of immune checkpoint inhibitors. Cancer Lett 2021; 509:53-62. [PMID: 33845122 DOI: 10.1016/j.canlet.2021.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/06/2021] [Accepted: 04/05/2021] [Indexed: 12/14/2022]
Abstract
Accumulating evidence suggests that the intestinal microbiota is associated with the antitumor efficacy of immune checkpoint inhibitors (ICIs) and the occurrence of immune-related adverse events (irAEs) following ICI treatment. However, the mechanisms underlying these interactions remain unclear. Recent technological advances have allowed more extensive investigation into the interplay between the intestinal microbiota and the tumor immune microenvironment. Breakthroughs by two research groups revealed that Bifidobacterium enhanced the efficacy of ICIs via the stimulator of interferon genes (STING) and adenosine 2A receptor (A2AR) signaling pathways, highlighting the molecular mechanisms through which the intestinal microbiota modulates immunotherapy. In this review, we summarize recent findings related to the potential role and mechanisms of the gut microbiota in ICI therapy, available microbiota-targeting strategies, and ongoing clinical trials. Further we discuss the associated challenges that remain in this field of research. The current review aims to evaluate the potential of the intestinal microbiota in maximizing the antitumor efficacy of ICIs while minimizing their toxic effects and guiding the development of more specific treatment regimens.
Collapse
Affiliation(s)
- Baohua Luo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yongbin Zhang
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Caiqin Zhang
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xiaoqiu Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
116
|
Ochi N, Ichihara E, Takigawa N, Harada D, Inoue K, Shibayama T, Hosokawa S, Kishino D, Harita S, Oda N, Hara N, Hotta K, Maeda Y, Kiura K. The effects of antibiotics on the efficacy of immune checkpoint inhibitors in patients with non-small-cell lung cancer differ based on PD-L1 expression. Eur J Cancer 2021; 149:73-81. [PMID: 33838391 DOI: 10.1016/j.ejca.2021.02.040] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/22/2021] [Accepted: 02/24/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are essential for treatment of various malignancies, including non-small-cell lung cancer (NSCLC). Recently, several studies have shown that the gut microbiome plays an important role in ICI treatment of solid cancers, and antibiotic (ATB) use had a negative impact on the outcomes of ICI treatment via dysbiosis in the gut. However, whether this is applicable to NSCLC remains unclear. The impact of ATBs based on PD-L1 expression also remains unclear. METHODS We retrospectively reviewed the medical records of patients with NSCLC who received ICI monotherapy (anti-PD-1 or anti-PD-L1 antibody) at nine institutions from December 2015 to May 2018. Outcomes with use of ATBs during the 2 months before or a month after initiation of ICI treatment, including progression-free survival (PFS) and overall survival (OS), were investigated using the Kaplan-Meier method. Multivariate analysis was also conducted using a Cox proportional hazards model. RESULTS A total of 531 patients were included in this study, among whom 98 (18.5%) received ATBs before or after ICI treatment. ATB use was significantly associated with a shorter median OS (11.7 months in the ATB group vs. 16.1 months in the non-ATB group; p = 0.028), whereas the difference in PFS was not significant (3.5 months in both the groups; p = 0.287). We next investigated the association based on PD-L1 expression in the 265 patients for whom PD-L1 expression was determined. There was no significant difference in the median OS or PFS between patients with NSCLC and PD-L1 expression <50% receiving ATBs and those not receiving ATBs (PFS: 3.3 vs. 2.8 months, p = 0.88; OS: 9.5 vs. 17.1 months, p = 0.24). Conversely, patients with NSCLC and PD-L1 expression ≥50% receiving ATBs showed significantly shorter median PFS and OS (PFS: 4.2 vs. 9.4 months, p = 0.012; OS: 11.9 vs. 28.4 months, p = 0.011). The impact of ATBs in patients with NSCLC and PD-L1 expression ≥50% was more significant than that in the entire cohort. CONCLUSIONS Our results indicate that the impact of ATB use on the efficacy of ICIs differed based on PD-L1 expression in patients with advanced NSCLC. A negative impact of ATB use was found in patients with NSCLC and PD-L1 expression ≥50% but not in those with PD-L1 expression <50%.
Collapse
Affiliation(s)
- Nobuaki Ochi
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Eiki Ichihara
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Japan.
| | - Nagio Takigawa
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Daijiro Harada
- Department of Thoracic Oncology, National Hospital Organization Shikoku Cancer Center, Japan
| | - Koji Inoue
- Department of Respiratory Medicine, Ehime Prefectural Central Hospital, Japan
| | - Takuo Shibayama
- Department of Respiratory Medicine, National Hospital Organization Okayama Medical Center, Japan
| | - Shinobu Hosokawa
- Department of Respiratory Medicine, Japanese Red Cross Okayama Hospital, Japan
| | - Daizo Kishino
- Department of Respiratory Medicine, Himeji Red Cross Hospital, Japan
| | - Shingo Harita
- Department of Internal Medicine, Okayama Saiseikai General Hospital, Japan
| | - Naohiro Oda
- Department of Internal Medicine, Fukuyama City Hospital, Japan
| | - Naofumi Hara
- Department of Hematology, Oncology and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Japan
| | - Katsuyuki Hotta
- Center for Innovative Clinical Medicine, Okayama University Hospital, Japan
| | - Yoshinobu Maeda
- Department of Hematology, Oncology and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Japan
| | - Katsuyuki Kiura
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Japan
| |
Collapse
|
117
|
Sipos A, Ujlaki G, Mikó E, Maka E, Szabó J, Uray K, Krasznai Z, Bai P. The role of the microbiome in ovarian cancer: mechanistic insights into oncobiosis and to bacterial metabolite signaling. Mol Med 2021; 27:33. [PMID: 33794773 PMCID: PMC8017782 DOI: 10.1186/s10020-021-00295-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer is characterized by dysbiosis, referred to as oncobiosis in neoplastic diseases. In ovarian cancer, oncobiosis was identified in numerous compartments, including the tumor tissue itself, the upper and lower female genital tract, serum, peritoneum, and the intestines. Colonization was linked to Gram-negative bacteria with high inflammatory potential. Local inflammation probably participates in the initiation and continuation of carcinogenesis. Furthermore, local bacterial colonies in the peritoneum may facilitate metastasis formation in ovarian cancer. Vaginal infections (e.g. Neisseria gonorrhoeae or Chlamydia trachomatis) increase the risk of developing ovarian cancer. Bacterial metabolites, produced by the healthy eubiome or the oncobiome, may exert autocrine, paracrine, and hormone-like effects, as was evidenced in breast cancer or pancreas adenocarcinoma. We discuss the possible involvement of lipopolysaccharides, lysophosphatides and tryptophan metabolites, as well as, short-chain fatty acids, secondary bile acids and polyamines in the carcinogenesis of ovarian cancer. We discuss the applicability of nutrients, antibiotics, and probiotics to harness the microbiome and support ovarian cancer therapy. The oncobiome and the most likely bacterial metabolites play vital roles in mediating the effectiveness of chemotherapy. Finally, we discuss the potential of oncobiotic changes as biomarkers for the diagnosis of ovarian cancer and microbial metabolites as possible adjuvant agents in therapy.
Collapse
Affiliation(s)
- Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Gyula Ujlaki
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Edit Mikó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Eszter Maka
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Debrecen, Egyetem tér 1, Debrecen, 4032, Hungary
| | - Judit Szabó
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Karen Uray
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Zoárd Krasznai
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Debrecen, Egyetem tér 1, Debrecen, 4032, Hungary
| | - Péter Bai
- Department of 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
|
118
|
Thinking Small: Small Molecules as Potential Synergistic Adjuncts to Checkpoint Inhibition in Melanoma. Int J Mol Sci 2021; 22:ijms22063228. [PMID: 33810078 PMCID: PMC8005112 DOI: 10.3390/ijms22063228] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/17/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022] Open
Abstract
Metastatic melanoma remains the deadliest form of skin cancer. Immune checkpoint inhibition (ICI) immunotherapy has defined a new age in melanoma treatment, but responses remain inconsistent and some patients develop treatment resistance. The myriad of newly developed small molecular (SM) inhibitors of specific effector targets now affords a plethora of opportunities to increase therapeutic responses, even in resistant melanoma. In this review, we will discuss the multitude of SM classes currently under investigation, current and prospective clinical combinations of ICI and SM therapies, and their potential for synergism in melanoma eradication based on established mechanisms of immunotherapy resistance.
Collapse
|
119
|
Targeting the Gut Microbiome to Mitigate Immunotherapy-Induced Colitis in Cancer. Trends Cancer 2021; 7:583-593. [PMID: 33741313 DOI: 10.1016/j.trecan.2021.02.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 12/14/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have been a transformational advance in cancer therapy in the past decade. However, ICIs can produce immune-related adverse effects (irAEs), which can lead to both morbidity and premature termination of therapy. Recent studies suggest that the gut microbiota and its metabolites affect ICI efficacy and toxicity. Herein, we review such evidence in the context of ICI-induced colitis. In particular, the short-chain fatty acid butyrate, a microbial metabolite, has known protective effects on the intestine. We discuss how the use of dietary prebiotics, which can be metabolized by bacteria to produce butyrate, can be an intriguing new investigational approach to prevent ICI-associated colitis and lead to improved patient outcomes.
Collapse
|
120
|
Salas-Benito D, Pérez-Gracia JL, Ponz-Sarvisé M, Rodriguez-Ruiz ME, Martínez-Forero I, Castañón E, López-Picazo JM, Sanmamed MF, Melero I. Paradigms on Immunotherapy Combinations with Chemotherapy. Cancer Discov 2021; 11:1353-1367. [PMID: 33712487 DOI: 10.1158/2159-8290.cd-20-1312] [Citation(s) in RCA: 189] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/21/2020] [Accepted: 01/04/2021] [Indexed: 11/16/2022]
Abstract
Checkpoint inhibitors are being added to standard-of-care chemotherapy in multiple clinical trials. Success has been reported in non-small and small cell lung carcinomas and urothelial, head and neck, gastric, and esophageal cancers, and promising results are already available in triple-negative breast and pancreatic malignancies. The potential mechanisms of synergy include immunogenic tumor cell death, antiangiogenesis, selective depletion of myeloid immunosuppressive cells, and lymphopenia, which reduces regulatory T cells and makes room for proliferation of effector T cells. However, chemotherapy regimens have not been optimized for such combinations, perhaps explaining some recent clinical trial disappointments. Approaches to make the most of chemoimmunotherapy include neoadjuvant and adjuvant schemes.Significance: Immunotherapy of cancer based on PD-1/PD-L1 blockade has prompted a revolution in cancer clinical management. Evidence in phase III clinical trials already supports combinations of immunotherapy with standard-of-care chemotherapy for a number of malignant diseases. This review focuses on such evidence and provides an overview of the potential synergistic mechanisms of action and the opportunities to optimize chemoimmunotherapy regimens.
Collapse
Affiliation(s)
- Diego Salas-Benito
- Oncology Department, Clínica Universidad de Navarra, Pamplona, Spain. .,Clinical Trials Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | - José L Pérez-Gracia
- Oncology Department, Clínica Universidad de Navarra, Pamplona, Spain.,Clinical Trials Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | - Mariano Ponz-Sarvisé
- Oncology Department, Clínica Universidad de Navarra, Pamplona, Spain.,Clinical Trials Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | - María E Rodriguez-Ruiz
- Oncology Department, Clínica Universidad de Navarra, Pamplona, Spain.,Clinical Trials Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | | | - Eduardo Castañón
- Oncology Department, Clínica Universidad de Navarra, Pamplona, Spain.,Clinical Trials Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | - José M López-Picazo
- Oncology Department, Clínica Universidad de Navarra, Pamplona, Spain.,Clinical Trials Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | - Miguel F Sanmamed
- Oncology Department, Clínica Universidad de Navarra, Pamplona, Spain.,Clinical Trials Unit, Clínica Universidad de Navarra, Pamplona, Spain.,Center for Medical Applied Research (CIMA), Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Biomedical Research Network in Oncology (CIBERONC), Pamplona, Spain
| | - Ignacio Melero
- Clinical Trials Unit, Clínica Universidad de Navarra, Pamplona, Spain. .,Center for Medical Applied Research (CIMA), Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Biomedical Research Network in Oncology (CIBERONC), Pamplona, Spain.,Immunology Department, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
121
|
Hopkins AM, Kichenadasse G, Abuhelwa AY, McKinnon RA, Rowland A, Sorich MJ. Value of the Lung Immune Prognostic Index in Patients with Non-Small Cell Lung Cancer Initiating First-Line Atezolizumab Combination Therapy: Subgroup Analysis of the IMPOWER150 Trial. Cancers (Basel) 2021; 13:cancers13051176. [PMID: 33803256 PMCID: PMC7967121 DOI: 10.3390/cancers13051176] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 12/16/2022] Open
Abstract
The lung immune prognostic index (LIPI) is proposed to differentiate prognosis and treatment benefit from immune checkpoint inhibitors (ICIs) in advanced non-small cell lung cancer (NSCLC). There is minimal information on the predictive importance with first-line, combination ICI approaches. In post-hoc analysis of IMpower150, Cox-proportional hazard analysis assessed the association between LIPI groups and overall survival (OS)/progression free survival (PFS). IMpower150 involved chemotherapy-naïve, metastatic non-squamous NSCLC participants randomized atezolizumab-carboplatin-paclitaxel (ACP), bevacizumab-carboplatin-paclitaxel (BCP), or atezolizumab-BCP (ABCP). Good (0 factors), intermediate (1 factor), and poor LIPI (2 factors) were defined via derived neutrophil-to-lymphocyte ratio >3, and lactate dehydrogenase >upper limit of normal. Of 1148 participants, 548 had good, 479 intermediate, and 121 poor LIPI. In 385 participants randomised ABCP, a significant association between LIPI and OS (HR (95%CI): intermediate LIPI = 2.16 (1.47-3.18), poor LIPI = 5.28 (3.20-8.69), p < 0.001) and PFS (HR (95%CI): intermediate LIPI = 1.47 (1.11-1.95), poor LIPI = 3.02 (2.03-4.50), p < 0.001) was identified. Median OS was 24, 16, and 7 months for good, intermediate, and poor LIPI, respectively. ACP associations were similar. Relative OS treatment effect (HR 95%CI) of ABCP vs. BCP was 0.78 (0.53-1.15), 0.67 (0.49-0.91), and 0.87 (0.51-1.47) for the good, intermediate, and poor LIPI groups, respectively (P(interaction) = 0.66), with no benefit in median OS observed in the poor LIPI group. LIPI identified subgroups with significantly different survival following ABCP and ACP initiation for chemotherapy-naïve, metastatic non-squamous NSCLC. There was insufficient evidence that LIPI identifies patients unlikely to benefit from ABCP treatment.
Collapse
Affiliation(s)
- Ashley M. Hopkins
- College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (G.K.); (A.Y.A.); (R.A.M.); (A.R.); (M.J.S.)
- Correspondence: ; Tel.: +61-8-8201-5647
| | - Ganessan Kichenadasse
- College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (G.K.); (A.Y.A.); (R.A.M.); (A.R.); (M.J.S.)
- Department of Medical Oncology, Flinders Centre for Innovation in Cancer, Flinders Medical Centre, Bedford Park, SA 5042, Australia
| | - Ahmad Y. Abuhelwa
- College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (G.K.); (A.Y.A.); (R.A.M.); (A.R.); (M.J.S.)
| | - Ross A. McKinnon
- College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (G.K.); (A.Y.A.); (R.A.M.); (A.R.); (M.J.S.)
| | - Andrew Rowland
- College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (G.K.); (A.Y.A.); (R.A.M.); (A.R.); (M.J.S.)
| | - Michael J. Sorich
- College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (G.K.); (A.Y.A.); (R.A.M.); (A.R.); (M.J.S.)
| |
Collapse
|
122
|
Food, Nutrition, Physical Activity and Microbiota: Which Impact on Lung Cancer? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18052399. [PMID: 33804536 PMCID: PMC7967729 DOI: 10.3390/ijerph18052399] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/11/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023]
Abstract
Lung cancer still represents the leading cause of cancer-related death, globally. Likewise, malnutrition and inactivity represent a major risk for loss of functional pulmonary capacities influencing overall lung cancer severity. Therefore, the adhesion to an appropriate health lifestyle is crucial in the management of lung cancer patients despite the subtype of cancer. This review aims to summarize the available knowledge about dietary approaches as well as physical activity as the major factors that decrease the risk towards lung cancer, and improve the response to therapies. We discuss the most significant dietary schemes positively associated to body composition and prognosis of lung cancer and the main molecular processes regulated by specific diet schemes, functional foods and physical activity, i.e., inflammation and oxidative stress. Finally, we report evidence demonstrating that dysbiosis of lung and/or gut microbiome, as well as their interconnection (the gut–lung axis), are strictly related to dietary patterns and regular physical activity playing a key role in lung cancer formation and progression, opening to the avenue of modulating the microbiome as coadjuvant therapy. Altogether, the evidence reported in this review highlights the necessity to consider non-pharmacological interventions (nutrition and physical activity) as effective adjunctive strategies in the management of lung cancer.
Collapse
|
123
|
The Lung Microbiome: A Central Mediator of Host Inflammation and Metabolism in Lung Cancer Patients? Cancers (Basel) 2020; 13:cancers13010013. [PMID: 33375062 PMCID: PMC7792810 DOI: 10.3390/cancers13010013] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/23/2020] [Accepted: 12/09/2020] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Lung cancer is the major cause of cancer related deaths in the world. New therapies have improved outcomes. Unfortunately, overall 5 year survival is ~20%. Therefore, better understanding of tumor biology and the microenvironment may lead to new therapeutic targets. The lung microbiome has recently emerged as a major mediator of host inflammation and pathogenesis. Understanding how the lung microbiota exerts its effects on lung cancer and the tumor microenvironment will allow for novel development of therapies. Abstract Lung cancer is the leading cause of cancer-related death. Over the past 5–10 years lung cancer outcomes have significantly improved in part due to better treatment options including immunotherapy and molecularly targeted agents. Unfortunately, the majority of lung cancer patients do not enjoy durable responses to these new treatments. Seminal research demonstrated the importance of the gut microbiome in dictating responses to immunotherapy in melanoma patients. However, little is known regarding how other sites of microbiota in the human body affect tumorigenesis and treatment responses. The lungs were traditionally thought to be a sterile environment; however, recent research demonstrated that the lung contains its own dynamic microbiota that can influence disease and pathophysiology. Few studies have explored the role of the lung microbiome in lung cancer biology. In this review article, we discuss the links between the lung microbiota and cancer, with particular focus on immune responses, metabolism and strategies to target the lung microbiome for cancer prevention.
Collapse
|
124
|
Pathak R, Pharaon RR, Mohanty A, Villaflor VM, Salgia R, Massarelli E. Acquired Resistance to PD-1/PD-L1 Blockade in Lung Cancer: Mechanisms and Patterns of Failure. Cancers (Basel) 2020; 12:cancers12123851. [PMID: 33419311 PMCID: PMC7767234 DOI: 10.3390/cancers12123851] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 02/08/2023] Open
Abstract
Simple Summary Acquired resistance to immunotherapy is an emerging issue, especially as the indications for immunotherapy expand to many different solid tumor malignancies. This review attempts to summarize the mechanisms and clinical outcomes of acquired resistance in non-small-cell lung cancers and explores future directions for research to understand and address this important question. Abstract Immunotherapy is now the preferred treatment for most lung cancer patients. It is used to treat unresectable stage III non-small-cell lung cancer and is the first-line therapy for non-oncogene-driven advanced/metastatic non-small-cell lung cancer patients (either alone or in combination with chemotherapy). Unfortunately, most patients that respond initially to immunotherapy develop resistance over time, thus limiting the durability of immunotherapy. A better understanding of the mechanisms of acquired resistance is urgently needed to expand the benefit of immunotherapy in lung cancer patients. This review aims to summarize the mechanisms and clinical outcomes of acquired resistance of anti-PD-1/PD-L1 therapies in non-small-cell lung cancer patients.
Collapse
|
125
|
Cren PY, Bertrand N, Le Deley MC, Génin M, Mortier L, Odou P, Penel N, Chazard E. Is the survival of patients treated with ipilimumab affected by antibiotics? An analysis of 1585 patients from the French National hospital discharge summary database (PMSI). Oncoimmunology 2020; 9:1846914. [PMID: 33299658 PMCID: PMC7714497 DOI: 10.1080/2162402x.2020.1846914] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background: The gut microbiota has a key role in the regulation of the immune system. Disruption of the gut microbiota’s composition by antibiotics might significantly affect the efficacy of immune checkpoint inhibitors. In a study of patients treated with ipilimumab, we sought to assess the relationship between overall survival and in-hospital antibiotic administration. Methods: Patients having been treated with ipilimumab between January 2012 and November 2014 were selected from the French National Hospital Discharge Summary Database. Exposure to antibiotics was defined as the presence of a hospital stay with a documented systemic bacterial infection in the 2 months before or the month after initiation of the patient’s first ever course of ipilimumab. The primary outcome was overall survival. Results: We studied 43,124 hospital stays involving 1585 patients from 97 centers. All patients had received ipilimumab monotherapy for advanced melanoma. Overall, 117 of the 1585 patients (7.4%) were documented as having received systemic antibiotic therapy in hospital during the defined exposure period. The median overall survival time was shorter in patients with infection (6.3 months, vs. 15.4 months in patients without an infection; hazard ratio (HR) = 1.88, 95% confidence interval [1.46; 2.43], p = 10−6). In a multivariate analysis adjusted for covariates, infection was still significantly associated with overall survival (HR = 1.68, [1.30; 2.18], p = 10−5). Conclusions: In patients treated with ipilimumab for advanced melanoma, infection, and antibiotic administration in hospital at around the time of the patient’s first ever course of ipilimumab appears to be associated with significantly lower clinical benefit.
Collapse
Affiliation(s)
- Pierre-Yves Cren
- CERIM, ULR 2694 METRICS, Univ. Lille, CHU Lille, Lille, France.,Methodology and Biostatistics Unit, Centre Oscar Lambret, Lille, France
| | | | - Marie-Cécile Le Deley
- Methodology and Biostatistics Unit, Centre Oscar Lambret, Lille, France.,CESP, INSERM, Paris-Saclay University, Paris-Sud University, UVSQ, Villejuif, France
| | - Michaël Génin
- CERIM, ULR 2694 METRICS, Univ. Lille, CHU Lille, Lille, France
| | - Laurent Mortier
- Clinique de Dermatologie, Unit#xE9; d#x27;Onco-Dermatologie, U1189, INSERM, Univ. Lille, CHU Lille, Lille, France
| | - Pascal Odou
- ULR 7365 GRITA, Univ. Lille, CHU Lille, Lille, France
| | - Nicolas Penel
- Department of Medical Oncology, Centre Oscar Lambret and Univ. Lille, Lille, France
| | | |
Collapse
|
126
|
Tonneau M, Elkrief A, Pasquier D, Paz Del Socorro T, Chamaillard M, Bahig H, Routy B. The role of the gut microbiome on radiation therapy efficacy and gastrointestinal complications: A systematic review. Radiother Oncol 2020; 156:1-9. [PMID: 33137398 DOI: 10.1016/j.radonc.2020.10.033] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/30/2020] [Accepted: 10/22/2020] [Indexed: 02/08/2023]
Abstract
Radiation therapy (RT) is an essential component of therapy either curative or palliative armamentarium in oncology, but its efficacy varies considerably among patients through many extrinsic and intrinsic mechanisms of the tumour, which are beginning to be better understood. Recent studies have shown that the gut microbiome represents a key factor in the modulation of the systemic immune response and consequently on patients' outcome. Moreover, the emergence of biomarkers that are derived from the gut microbiota has fuelled the development of adjuvant strategies for patients treated with immunotherapy in combination or not with RT. Despite progress in development of more precise radiotherapy techniques, almost all patients undergoing RT to the abdomen, pelvis, or rectum develop acute adverse events as a consequence of several dose-limiting parameters such as the location of irradiation that may subsequently damage normal tissue including the intestinal epithelium. Several lines of evidence in preclinical models identified that vancomycin improves RT-induced gastrointestinal toxicities such as diarrhea and oral mucositis. In order to gain further insight into this rapidly evolving field, we have systematically reviewed the studies that have described how the gut microbiome may directly or indirectly modulate RT efficacy and its gastro-intestinal toxicities. Lastly, we outline current knowledge gaps and discuss potentially more satisfactory therapeutic options to restore the functionality of the gut microbiome of patients treated with RT.
Collapse
Affiliation(s)
- Marion Tonneau
- Département universitaire de radiothérapie, Centre Oscar Lambret, Lille, France
| | - Arielle Elkrief
- Centre de recherche de l'Université de Montréal, (CRCHUM), Montréal, QC, Canada
| | - David Pasquier
- Département universitaire de radiothérapie, Centre Oscar Lambret, Lille, France; CRIStAL UMR 9189, Lille University, France; Univ. Lille, Inserm, U1003 - PHYCEL - Physiologie Cellulaire, F-59000, Lille, France
| | | | - Mathias Chamaillard
- Univ. Lille, Inserm, U1003 - PHYCEL - Physiologie Cellulaire, F-59000, Lille, France
| | - Houda Bahig
- Centre de recherche de l'Université de Montréal, (CRCHUM), Montréal, QC, Canada; Centre hospitalier de l'Université de Montréal, (CHUM), Montréal, QC, Canada
| | - Bertrand Routy
- Centre de recherche de l'Université de Montréal, (CRCHUM), Montréal, QC, Canada; Centre hospitalier de l'Université de Montréal, (CHUM), Montréal, QC, Canada.
| |
Collapse
|
127
|
Medjebar S, Truntzer C, Perrichet A, Limagne E, Fumet JD, Richard C, Elkrief A, Routy B, Rébé C, Ghiringhelli F. Angiotensin-converting enzyme (ACE) inhibitor prescription affects non-small-cell lung cancer (NSCLC) patients response to PD-1/PD-L1 immune checkpoint blockers. Oncoimmunology 2020; 9:1836766. [PMID: 33178495 PMCID: PMC7595630 DOI: 10.1080/2162402x.2020.1836766] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 10/06/2020] [Accepted: 10/10/2020] [Indexed: 12/15/2022] Open
Abstract
Angiotensin-converting enzyme (ACE) inhibitors are frequently used to treat hypertension and congestive heart failure. Preclinical data show that ACE plays a role on both innate and adaptive immune responses. Since interactions between ACE inhibitors and immune checkpoint inhibitors (ICIs) have not been reported, the aim of this study is to investigate the influence of ACE inhibitors on non-small cell lung cancer (NSCLC) patients treated with programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) inhibitors. We conducted a retrospective cohort analysis of NSCLC patients treated with PD-1/PD-L1 inhibitors. Clinical and co-medication data as well as tumor biopsies were collected. Groups were defined according to patients' co-medications at the time of ICI initiation. Among the 178 patients included, 22 (13.1%) received ACE inhibitors. While baseline characteristics were similar in both groups, ACE inhibitors group had a shorter median PFS (Progression-Free Survival) compared to the control group: 1.97 vs. 2.56 months, p = .01 (HR = 1.8 CI95% 1.1-2.8). Using CIBERSORT, RNA sequencing suggested that tumors from the ACE inhibitors group had less M1 macrophages, activated mast cells, NK cells and memory activated T cells, thus suggesting an immunosuppressed state. In vitro, the ACE inhibitor, captopril, induced M2 marker at the cell surface of monocytes engaged into M1 differentiation. Thus, ACE inhibitors prescription concomitant to PD-1/PD-L1 inhibitors treatment seems to be associated with impaired outcome and with a tumor immunosuppressed state in patients with advanced NSCLC. These results should be validated in larger prospective cohorts.
Collapse
Affiliation(s)
- Soleine Medjebar
- Department of Medical Oncology, GF Leclerc Centre, Dijon, France
- Platform of Transfer in Cancer Biology, GF Leclerc Centre, Dijon, France
| | - Caroline Truntzer
- Platform of Transfer in Cancer Biology, GF Leclerc Centre, Dijon, France
- University of Bourgogne-Franche-Comté, Dijon, France
- Genetic and Immunology Medical Institute (GIMI), Dijon, France
- INSERM UMR1231, Dijon, France
| | - Anaïs Perrichet
- Platform of Transfer in Cancer Biology, GF Leclerc Centre, Dijon, France
- University of Bourgogne-Franche-Comté, Dijon, France
- INSERM UMR1231, Dijon, France
| | - Emeric Limagne
- Platform of Transfer in Cancer Biology, GF Leclerc Centre, Dijon, France
- University of Bourgogne-Franche-Comté, Dijon, France
- INSERM UMR1231, Dijon, France
| | - Jean-David Fumet
- Department of Medical Oncology, GF Leclerc Centre, Dijon, France
- Platform of Transfer in Cancer Biology, GF Leclerc Centre, Dijon, France
- University of Bourgogne-Franche-Comté, Dijon, France
| | - Corentin Richard
- Platform of Transfer in Cancer Biology, GF Leclerc Centre, Dijon, France
- University of Bourgogne-Franche-Comté, Dijon, France
| | - Arielle Elkrief
- Research Centre for the University of Montréal (CRCHUM), Montréal. Hematology-Oncology Division, Department of Medicine, University of Montreal Healthcare Centre (CHUM), Montreal, Canada
| | - Bertrand Routy
- Research Centre for the University of Montréal (CRCHUM), Montréal. Hematology-Oncology Division, Department of Medicine, University of Montreal Healthcare Centre (CHUM), Montreal, Canada
| | - Cédric Rébé
- Platform of Transfer in Cancer Biology, GF Leclerc Centre, Dijon, France
- University of Bourgogne-Franche-Comté, Dijon, France
- INSERM UMR1231, Dijon, France
| | - François Ghiringhelli
- Department of Medical Oncology, GF Leclerc Centre, Dijon, France
- Platform of Transfer in Cancer Biology, GF Leclerc Centre, Dijon, France
- University of Bourgogne-Franche-Comté, Dijon, France
- Genetic and Immunology Medical Institute (GIMI), Dijon, France
- INSERM UMR1231, Dijon, France
| |
Collapse
|
128
|
Sári Z, Mikó E, Kovács T, Boratkó A, Ujlaki G, Jankó L, Kiss B, Uray K, Bai P. Indoxylsulfate, a Metabolite of the Microbiome, Has Cytostatic Effects in Breast Cancer via Activation of AHR and PXR Receptors and Induction of Oxidative Stress. Cancers (Basel) 2020; 12:E2915. [PMID: 33050543 PMCID: PMC7599465 DOI: 10.3390/cancers12102915] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 09/29/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023] Open
Abstract
Changes to bacterial metabolite-elicited signaling, in oncobiosis associated with breast cancer, plays a role in facilitating the progression of the disease. We show that indoxyl-sulfate (IS), a tryptophan metabolite, has cytostatic properties in models of breast cancer. IS supplementation, in concentrations corresponding to the human serum reference range, suppressed tumor infiltration to the surrounding tissues and metastasis formation in a murine model of breast cancer. In cellular models, IS suppressed NRF2 and induced iNOS, leading to induction of oxidative and nitrosative stress, and, consequently, reduction of cell proliferation; enhanced oxidative and nitrosative stress are crucial in the subsequent cytostasis. IS also suppressed epithelial-to-mesenchymal transition vital for suppressing cellular movement and diapedesis. Furthermore, IS rendered cells hypometabolic, leading to a reduction in aldehyde-dehydrogenase positive cells. Pharmacological inhibition of the pregnane-X receptor using CH223191 and the aryl-hydrocarbon receptor using ketoconazole diminished the IS-elicited effects, suggesting that these receptors were the major receptors of IS in these models. Finally, we showed that increased expression of the human enzymes that form IS (Cyp2E1, Sult1A1, and Sult1A2) is associated with better survival in breast cancer, an effect that is lost in triple negative cases. Taken together, IS, similar to indolepropionic acid (another tryptophan metabolite), has cytostatic properties and higher expression of the metabolic machinery responsible for the formation of IS supports survival in breast cancer.
Collapse
Affiliation(s)
- Zsanett Sári
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1., 4032 Debrecen, Hungary; (Z.S.); (E.M.); (T.K.); (A.B.); (G.U.); (L.J.); (K.U.)
| | - Edit Mikó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1., 4032 Debrecen, Hungary; (Z.S.); (E.M.); (T.K.); (A.B.); (G.U.); (L.J.); (K.U.)
- MTA-DE Lendület Laboratory of Cellular Metabolism, 4032 Debrecen, Hungary
| | - Tünde Kovács
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1., 4032 Debrecen, Hungary; (Z.S.); (E.M.); (T.K.); (A.B.); (G.U.); (L.J.); (K.U.)
| | - Anita Boratkó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1., 4032 Debrecen, Hungary; (Z.S.); (E.M.); (T.K.); (A.B.); (G.U.); (L.J.); (K.U.)
| | - Gyula Ujlaki
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1., 4032 Debrecen, Hungary; (Z.S.); (E.M.); (T.K.); (A.B.); (G.U.); (L.J.); (K.U.)
| | - Laura Jankó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1., 4032 Debrecen, Hungary; (Z.S.); (E.M.); (T.K.); (A.B.); (G.U.); (L.J.); (K.U.)
| | - Borbála Kiss
- Department of Oncology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Karen Uray
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1., 4032 Debrecen, Hungary; (Z.S.); (E.M.); (T.K.); (A.B.); (G.U.); (L.J.); (K.U.)
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1., 4032 Debrecen, Hungary; (Z.S.); (E.M.); (T.K.); (A.B.); (G.U.); (L.J.); (K.U.)
- MTA-DE Lendület Laboratory of Cellular Metabolism, 4032 Debrecen, Hungary
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
129
|
Wang J, Yang HR, Wang DJ, Wang XX. Association between the gut microbiota and patient responses to cancer immune checkpoint inhibitors. Oncol Lett 2020; 20:342. [PMID: 33123253 PMCID: PMC7583737 DOI: 10.3892/ol.2020.12205] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023] Open
Abstract
Studies are increasingly investigating the association between the gut microbiota and the outcomes of immunotherapy in patients with cancer. Notably, certain studies have demonstrated that the gut microbiota serves a key role in regulating a patient's response to immunotherapy. In the present review, the potential associations between the gut microbiota, and cancer, host immunity and cancer immunotherapy are reviewed. Furthermore, the effects of fecal microbiota transplantation, antibiotics, probiotics, prebiotics, synbiotics, components of traditional Chinese medicine and various lifestyle factors on the gut microbiota and cancer immunotherapy outcomes are discussed. Certain dominant bacterial groups in the context of cancer immunotherapy and certain effective methods for optimizing immunotherapy by regulating the gut microbiota have been identified. Further investigation may enable the rapid conversion of these discoveries into practical products and clinically applicable methods.
Collapse
Affiliation(s)
- Jian Wang
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, P.R. China
| | - Hong-Ru Yang
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, P.R. China
| | - Dai-Jie Wang
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, P.R. China
| | - Xing-Xia Wang
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
130
|
Lung Microbiome Differentially Impacts Survival of Patients with Non-Small Cell Lung Cancer Depending on Tumor Stroma Phenotype. Biomedicines 2020; 8:biomedicines8090349. [PMID: 32933105 PMCID: PMC7554830 DOI: 10.3390/biomedicines8090349] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 01/01/2023] Open
Abstract
The link between a lung tumor and the lung microbiome is a largely unexplored issue. To investigate the relationship between a lung microbiome and the phenotype of an inflammatory stromal infiltrate, we studied a cohort of 89 patients with non-small cell lung cancer. The microbiome was analyzed in tumor and adjacent normal tissue by 16S rRNA amplicon sequencing. Characterization of the tumor stroma was done using immunohistochemistry. We demonstrated that the bacterial load was higher in adjacent normal tissue than in a tumor (p = 0.0325) with similar patterns of taxonomic structure and alpha diversity. Lung adenocarcinomas did not differ in their alpha diversity from squamous cell carcinomas, although the content of Gram-positive bacteria increased significantly in the adenocarcinoma group (p = 0.0419). An analysis of an inflammatory infiltrate of tumor stroma showed a correlation of CD68, iNOS and FOXP3 with a histological type of tumor. For the first time we showed that high bacterial load in the tumor combined with increased iNOS expression is a favorable prognostic factor (HR = 0.1824; p = 0.0123), while high bacterial load combined with the increased number of FOXP3+ cells is a marker of poor prognosis (HR = 4.651; p = 0.0116). Thus, we established that bacterial load of the tumor has an opposite prognostic value depending on the status of local antitumor immunity.
Collapse
|
131
|
Lee KA, Shaw HM, Bataille V, Nathan P, Spector TD. Role of the gut microbiome for cancer patients receiving immunotherapy: Dietary and treatment implications. Eur J Cancer 2020; 138:149-155. [PMID: 32889369 DOI: 10.1016/j.ejca.2020.07.026] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 07/01/2020] [Accepted: 07/22/2020] [Indexed: 01/01/2023]
Abstract
Immune-checkpoint inhibitors (ICIs) have revolutionised the therapeutic landscape for multiple malignancies and the health of the gut microbiome (GM) is strongly linked with therapeutic responses to ICI. This review explores the implications of diet and medication on the GM for patients receiving ICI. Clinical trials are underway to explore the impact of factors such as faecal microbiota transfer, probiotics, prebiotics, bacteria consortia and a number of dietary interventions on patients receiving ICI. Randomised controlled trials are lacking, and inferences are currently based on short-term clinical and observational studies. Antibiotics should be avoided before ICI initiation, and depending on prospective data, future consideration may be given to temporary delay of initiation of non-urgent ICI if patient has had broad spectrum antibiotics within 1 month of planned treatment initiation. Proton pump inhibitor use should be discontinued when not clearly indicated and potential switch to a histamine H2-receptor antagonist considered. Patients should be advised to minimise animal meat intake and maximise plants, aiming to consume ≥30 plant types weekly. A high fibre intake (>30 g/day) has been seen to be beneficial in increasing the chance of ICI response. Fermented foods may have a beneficial effect on the GM and should be introduced where possible. Ideally, all patients should be referred to a nutritionist or dietician with knowledge of GM before commencing ICI.
Collapse
Affiliation(s)
- Karla 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.
| | - Heather M Shaw
- Department of Medical Oncology, Mount Vernon Hospital, Northwood, UK; Early Phase Trial Unit, Department of Medical Oncology, University College London Hospital, London, UK
| | - Veronique Bataille
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK; Department of Dermatology, Mount Vernon Hospital, Northwood, UK
| | - Paul Nathan
- Department of Medical Oncology, Mount Vernon Hospital, Northwood, UK
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| |
Collapse
|
132
|
Real-world outcomes treating patients with advanced cutaneous squamous cell carcinoma with immune checkpoint inhibitors (CPI). Br J Cancer 2020; 123:1535-1542. [PMID: 32868898 PMCID: PMC7653959 DOI: 10.1038/s41416-020-01044-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/23/2020] [Accepted: 07/07/2020] [Indexed: 12/17/2022] Open
Abstract
Background Immunotherapy has revolutionised the treatment of advanced cutaneous squamous cell carcinoma (cSCC). It is important to understand both safety and efficacy in a real-world and trial-ineligible cSCC population. We aimed to evaluate safety, efficacy and molecular insights among a broader cSCC population, including immunosuppressed patients, treated with immune checkpoint inhibitors (CPI). Methods We present a cohort of advanced cSCC patients (n = 61) treated from 2015 to 2020 evaluating the best overall response (BOR) (RECISTv1.1) to CPI therapy, immune-related adverse events (irAEs) and tumour mutational burden (TMB) to correlate with outcomes. A validated geriatric scoring index (CIRS-G) was utilised to assess comorbidities among patients ≥75. These data were compared with published clinical trial results among the broader cSCC population. Results BOR to CPI was lower among the entire cohort when compared with trial data (31.5 vs. 48%, P < 0.01), with higher rates of progression (59 vs. 16.5%, P < 0.01), regardless of immunosuppression history or age. Grade 3+ irAEs were more common among responders (P = 0.02), while pre-treatment lymphocyte count and TMB predicted response (P = 0.02). Conclusions We demonstrate comparatively lower response rates to CPI among real-world cSCC patients not explained by older age or immunosuppression history alone. Immune-related toxicity, absolute lymphocyte count and TMB predicted CPI response.
Collapse
|
133
|
Leonardi GC, Candido S, Falzone L, Spandidos DA, Libra M. Cutaneous melanoma and the immunotherapy revolution (Review). Int J Oncol 2020; 57:609-618. [PMID: 32582963 PMCID: PMC7384846 DOI: 10.3892/ijo.2020.5088] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022] Open
Abstract
In a relatively short period of time, treatment strategies for metastatic melanoma have radically changed leading to an unprecedented improvement in patient survival. In this period, immunotherapy options have evolved from cytokine‑based approaches to antibody‑mediated inhibition of immune checkpoints, cancer vaccines and pharmacological modulation of the melanoma microenvironment. Combination of immunotherapy strategies and the association of immune checkpoint inhibitors (ICIs) with BRAF V600 targeted therapy show encouraging results. The future of drug development in this field is promising. The comprehension of primary and acquired resistance mechanisms to ICIs and the dissection of melanoma immunobiology will be instrumental for the development of new treatment strategies and to improve clinical trial design. Moreover, biomarker discovery will help patient stratification and management during immunotherapy treatment. In this review, we summarize landmark clinical trials of immune checkpoint inhibitors in advanced melanoma and discuss the rational for immunotherapy combinations. Immunotherapy approaches at early stage of clinical development and recent advances in melanoma immunotherapy biomarker development are also discussed.
Collapse
Affiliation(s)
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, I-95123 Catania
| | - Luca Falzone
- Epidemiology Unit, IRCCS Istituto Nazionale Tumori 'Fondazione G. Pascale', I-80131 Naples, Italy
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 70013 Heraklion, Greece
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, I-95123 Catania
| |
Collapse
|
134
|
Sári Z, Mikó E, Kovács T, Jankó L, Csonka T, Lente G, Sebő É, Tóth J, Tóth D, Árkosy P, Boratkó A, Ujlaki G, Török M, Kovács I, Szabó J, Kiss B, Méhes G, Goedert JJ, Bai P. Indolepropionic Acid, a Metabolite of the Microbiome, Has Cytostatic Properties in Breast Cancer by Activating AHR and PXR Receptors and Inducing Oxidative Stress. Cancers (Basel) 2020; 12:E2411. [PMID: 32854297 PMCID: PMC7565149 DOI: 10.3390/cancers12092411] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 08/20/2020] [Indexed: 02/07/2023] Open
Abstract
Oncobiotic transformation of the gut microbiome may contribute to the risk of breast cancer. Recent studies have provided evidence that the microbiome secretes cytostatic metabolites that inhibit the proliferation, movement, and metastasis formation of cancer cells. In this study, we show that indolepropionic acid (IPA), a bacterial tryptophan metabolite, has cytostatic properties. IPA selectively targeted breast cancer cells, but it had no effects on non-transformed, primary fibroblasts. In cell-based and animal experiments, we showed that IPA supplementation reduced the proportions of cancer stem cells and the proliferation, movement, and metastasis formation of cancer cells. These were achieved through inhibiting epithelial-to-mesenchymal transition, inducing oxidative and nitrosative stress, and boosting antitumor immune response. Increased oxidative/nitrosative stress was due to the IPA-mediated downregulation of nuclear factor erythroid 2-related factor 2 (NRF2), upregulation of inducible nitric oxide synthase (iNOS), and enhanced mitochondrial reactive species production. Increased oxidative/nitrosative stress led to cytostasis and reductions in cancer cell stem-ness. IPA exerted its effects through aryl hydrocarbon receptor (AHR) and pregnane X receptor (PXR) receptors. A higher expression of PXR and AHR supported better survival in human breast cancer patients, highlighting the importance of IPA-elicited pathways in cytostasis in breast cancer. Furthermore, AHR activation and PXR expression related inversely to cancer cell proliferation level and to the stage and grade of the tumor. The fecal microbiome's capacity for IPA biosynthesis was suppressed in women newly diagnosed with breast cancer, especially with stage 0. Bacterial indole biosynthesis showed correlation with lymphocyte infiltration to tumors in humans. Taken together, we found that IPA is a cytostatic bacterial metabolite, the production of which is suppressed in human breast cancer. Bacterial metabolites, among them, IPA, have a pivotal role in regulating the progression but not the initiation of the disease.
Collapse
Affiliation(s)
- Zsanett Sári
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (E.M.); (T.K.); (L.J.); (G.L.); (A.B.); (G.U.)
| | - Edit Mikó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (E.M.); (T.K.); (L.J.); (G.L.); (A.B.); (G.U.)
- MTA-DE Lendület Laboratory of Cellular Metabolism, 4032 Debrecen, Hungary
| | - Tünde Kovács
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (E.M.); (T.K.); (L.J.); (G.L.); (A.B.); (G.U.)
| | - Laura Jankó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (E.M.); (T.K.); (L.J.); (G.L.); (A.B.); (G.U.)
| | - Tamás Csonka
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.C.); (G.M.)
| | - Gréta Lente
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (E.M.); (T.K.); (L.J.); (G.L.); (A.B.); (G.U.)
| | - Éva Sebő
- Kenézy Breast Center at Kenézy Gyula County Hospital, 4032 Debrecen, Hungary;
| | - Judit Tóth
- Department of Oncology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (J.T.); (P.Á.); (B.K.)
| | - Dezső Tóth
- Department of Surgery, Borsod-Abaúj-Zemplén County Hospital and University Teaching Hospital, 3526 Miskolc, Hungary;
| | - Péter Árkosy
- Department of Oncology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (J.T.); (P.Á.); (B.K.)
| | - Anita Boratkó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (E.M.); (T.K.); (L.J.); (G.L.); (A.B.); (G.U.)
| | - Gyula Ujlaki
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (E.M.); (T.K.); (L.J.); (G.L.); (A.B.); (G.U.)
| | - Miklós Török
- Department of Pathology at Kenézy Gyula County Hospital, 4032 Debrecen, Hungary; (M.T.); (I.K.)
| | - Ilona Kovács
- Department of Pathology at Kenézy Gyula County Hospital, 4032 Debrecen, Hungary; (M.T.); (I.K.)
| | - Judit Szabó
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Borbála Kiss
- Department of Oncology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (J.T.); (P.Á.); (B.K.)
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.C.); (G.M.)
| | - James J. Goedert
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20982, USA;
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (E.M.); (T.K.); (L.J.); (G.L.); (A.B.); (G.U.)
- MTA-DE Lendület Laboratory of Cellular Metabolism, 4032 Debrecen, Hungary
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
135
|
Seddiki N, Picard F, Dupaty L, Lévy Y, Godot V. The Potential of Immune Modulation in Therapeutic HIV-1 Vaccination. Vaccines (Basel) 2020; 8:vaccines8030419. [PMID: 32726934 PMCID: PMC7565497 DOI: 10.3390/vaccines8030419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 02/07/2023] Open
Abstract
We discuss here some of the key immunological elements that are at the crossroads and need to be combined to develop a potent therapeutic HIV-1 vaccine. Therapeutic vaccines have been commonly used to enhance and/or recall pre-existing HIV-1-specific cell-mediated immune responses aiming to suppress virus replication. The current success of immune checkpoint blockers in cancer therapy renders them very attractive to use in HIV-1 infected individuals with the objective to preserve the function of HIV-1-specific T cells from exhaustion and presumably target the persistent cellular reservoir. The major latest advances in our understanding of the mechanisms responsible for virus reactivation during therapy-suppressed individuals provide the scientific basis for future combinatorial therapeutic vaccine development.
Collapse
Affiliation(s)
- Nabila Seddiki
- Inserm, U955, Equipe 16, 94000 Créteil, France; (F.P.); (L.D.); (Y.L.); (V.G.)
- Faculté de médecine, Université Paris Est, 94000 Créteil, France
- Vaccine Research Institute (VRI), 94000 Créteil, France
- INSERM U955 Equipe 16, Université Paris-Est Créteil, Vaccine Research Institute (VRI), 51, Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
- Correspondence: ; Tel.: +33-01-4981-3902; Fax: +33-01-4981-3709
| | - Florence Picard
- Inserm, U955, Equipe 16, 94000 Créteil, France; (F.P.); (L.D.); (Y.L.); (V.G.)
- Vaccine Research Institute (VRI), 94000 Créteil, France
| | - Léa Dupaty
- Inserm, U955, Equipe 16, 94000 Créteil, France; (F.P.); (L.D.); (Y.L.); (V.G.)
- Vaccine Research Institute (VRI), 94000 Créteil, France
| | - Yves Lévy
- Inserm, U955, Equipe 16, 94000 Créteil, France; (F.P.); (L.D.); (Y.L.); (V.G.)
- Faculté de médecine, Université Paris Est, 94000 Créteil, France
- Vaccine Research Institute (VRI), 94000 Créteil, France
- AP-HP Hôpital H. Mondor—A. Chenevier, Service d’Immunologie clinique et maladies infectieuses, 94010 Créteil, France
| | - Véronique Godot
- Inserm, U955, Equipe 16, 94000 Créteil, France; (F.P.); (L.D.); (Y.L.); (V.G.)
- Faculté de médecine, Université Paris Est, 94000 Créteil, France
- Vaccine Research Institute (VRI), 94000 Créteil, France
| |
Collapse
|
136
|
Hakozaki T, Richard C, Elkrief A, Hosomi Y, Benlaïfaoui M, Mimpen I, Terrisse S, Derosa L, Zitvogel L, Routy B, Okuma Y. The Gut Microbiome Associates with Immune Checkpoint Inhibition Outcomes in Patients with Advanced Non-Small Cell Lung Cancer. Cancer Immunol Res 2020; 8:1243-1250. [PMID: 32847937 DOI: 10.1158/2326-6066.cir-20-0196] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/20/2020] [Accepted: 07/24/2020] [Indexed: 01/10/2023]
Abstract
The gut microbiome (GM) plays an important role in shaping systemic immune responses and influences immune checkpoint inhibitor (ICI) efficacy. Antibiotics worsen clinical outcomes in patients receiving ICI. However, whether GM profiling and baseline antibiotic can be a biomarker of ICI efficacy in advanced non-small cell lung cancer (NSCLC) remains unknown. We prospectively collected baseline (pre-ICI) fecal samples and clinical data of 70 Japanese patients suffering from advanced NSCLC and treated them with anti-PD-1/PD-L1 antibodies as a first-line or treatment-refractory therapy. We performed 16S rRNA V3-V4 sequencing of gene amplicons of fecal samples, and bacteria diversity and differential abundance analysis was performed. The clinical endpoints were objective response rate (ORR), progression-free survival (PFS), overall survival (OS), and immune-related adverse events (irAE). ORR was 34%, and median PFS and OS were 5.2 and 16.2 months, respectively. Patients who received pre-ICI antibiotic had lower alpha diversity at baseline and underrepresentation of Ruminococcaceae UCG 13 and Agathobacter When analyzing antibiotic-free patients, alpha diversity correlated with OS. In addition, Ruminococcaceae UCG 13 and Agathobacter were enriched in patients with favorable ORR and PFS >6 months. Ruminococcaceae UCG 13 was enriched in patients with OS >12 months. GM differences were observed between patients who experienced low- versus high-grade irAE. We demonstrated the negative influence of antibiotic on the GM composition and identified the bacteria repertoire in patients experiencing favorable responses to ICI.See articles by Tomita et al., p. 1236, and Peng et al., p. 1251.
Collapse
Affiliation(s)
- Taiki Hakozaki
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Bunkyo City, Tokyo, Japan
| | - Corentin Richard
- University of Montreal Research Center (CRCHUM), Montreal, Quebec, Canada
| | - Arielle Elkrief
- University of Montreal Research Center (CRCHUM), Montreal, Quebec, Canada.,Segal Cancer Centre, Jewish General Hospital, Department of Oncology, Montreal, Quebec, Canada
| | - Yukio Hosomi
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Bunkyo City, Tokyo, Japan
| | - Myriam Benlaïfaoui
- University of Montreal Research Center (CRCHUM), Montreal, Quebec, Canada
| | - Iris Mimpen
- University of Montreal Research Center (CRCHUM), Montreal, Quebec, Canada
| | - Safae Terrisse
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France.,Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Villejuif, France.,Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Lisa Derosa
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France.,Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Villejuif, France.,Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France.,Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Villejuif, France.,Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Bertrand Routy
- University of Montreal Research Center (CRCHUM), Montreal, Quebec, Canada. .,Division of Medicine, Department of Hemato-Oncology, University of Montreal Healthcare Center, Montreal, Quebec, Canada
| | - Yusuke Okuma
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Bunkyo City, Tokyo, Japan. .,Department of Thoracic Oncology, National Cancer Center Hospital, Chuo City, Tokyo, Japan
| |
Collapse
|
137
|
Can we harness the microbiota to enhance the efficacy of cancer immunotherapy? Nat Rev Immunol 2020; 20:522-528. [PMID: 32661409 DOI: 10.1038/s41577-020-0374-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2020] [Indexed: 12/17/2022]
Abstract
There is currently much interest in defining how the microbiota shapes immune responses in the context of cancer. Various studies in both mice and humans have associated particular commensal species with better (or worse) outcomes in different cancer types and following treatment with cancer immunotherapies. However, the mechanisms involved remain ill-defined and even controversial. In this Viewpoint, Nature Reviews Immunology has invited six eminent scientists in the field to share their thoughts on the key questions and challenges for the field.
Collapse
|
138
|
Concomitant Antibiotic Use and Survival in Urothelial Carcinoma Treated with Atezolizumab. Eur Urol 2020; 78:540-543. [PMID: 32660748 DOI: 10.1016/j.eururo.2020.06.061] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022]
Abstract
Antibiotic effects on the gut microbiota may negatively impact survival with immune checkpoint inhibitors (ICIs). However, there is minimal evidence regarding whether antibiotic impacts are specific to ICIs or impacts in urothelial carcinoma (UC). In a post hoc analysis of IMvigor210 (single-arm atezolizumab) and IMvigor211 (phase III randomised trial of atezolizumab vs chemotherapy), the association between antibiotic use and overall survival (OS) and progression-free survival (PFS) was assessed via Cox proportional hazard analysis. Antibiotic use was defined as any antibiotic administration between 30 d prior to and 30 d after treatment initiation. Antibiotic use was associated with worse OS (n = 847, hazard ratio or HR [95% confidence interval {CI}] = 1.44 [1.19-1.73]) and PFS (1.24 [1.05-1.46]) with atezolizumab, but not chemotherapy (n = 415, 1.15 [0.91-1.46] and 1.09 [0.88-1.36], respectively). In the randomised cohort of IMvigor211, the OS treatment effect (HR [95% CI]) of atezolizumab versus chemotherapy was 0.95 (95% CI 0.71-1.25) for antibiotic users, compared with 0.73 (0.60-0.88) for nonusers (p[interaction] = 0.1). Similar associations were noted in the PD-L1 IC2/3 population. In conclusion, antibiotic use was associated with worse survival outcomes in UC patients treated with atezolizumab. The study does not justify a change in antibiotic selection for infections; however antibiotic overuse occurs in cancer care and this needs to be evaluated for ICIs. PATIENT SUMMARY: In this report from clinical trials IMvigor210 and IMvigor211, it was demonstrated that antibiotic use is consistently associated with worse survival in patients with urothelial carcinoma treated with atezolizumab. No antibiotic association was observed in patients treated with chemotherapy, suggesting that antibiotics may specifically reduce the effectiveness of cancer immunotherapies. Future research will continue to explore the effect of antibiotics on other immune checkpoint inhibitors and confirm whether immune checkpoint inhibitors remain the treatment of choice in cancer patients requiring antibiotics.
Collapse
|
139
|
Moreira M, Pobel C, Epaillard N, Simonaggio A, Oudard S, Vano YA. Resistance to cancer immunotherapy in metastatic renal cell carcinoma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:454-471. [PMID: 35582435 PMCID: PMC8992500 DOI: 10.20517/cdr.2020.16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/06/2020] [Accepted: 05/29/2020] [Indexed: 12/17/2022]
Abstract
The prognosis of metastatic clear cell renal cell carcinoma (mccRCC) has changed dramatically over the years with the emergence of immune checkpoint inhibitors (ICI) used alone, or in combination with another ICI, or with vascular endothelial growth factor receptor tyrosine kinase inhibitor. Although major response rates have been observed with ICI, many patients do not respond, reflecting primary resistance, and durable responses remain exceptional, reflecting secondary resistance. Factors contributing to primary and acquired resistance are manifold, including patient-intrinsic factors, tumor cell-intrinsic factors and factors associated with the tumoral microenvironment (TME). While some mechanisms of resistance are common to several tumor types, others are specific to mccRCC. Predictive biomarkers and alternative strategies are needed to overcome this resistance. This review provides an overview of the major ICI resistance mechanisms, highlights the potential of the TME to induce resistance to ICI, and discusses the predictive biomarkers available to guide therapeutic choice.
Collapse
Affiliation(s)
- Marco Moreira
- Centre de Recherche des Cordeliers, INSERM, Université de Paris, Sorbonne Université, Paris F-75006, France.,Both authors contributed equally
| | - Cedric Pobel
- Department of Medical Oncology, European Hospital Georges Pompidou - APHP, Paris 75015, France.,University François Rabelais, Tours 37200, France.,Both authors contributed equally
| | - Nicolas Epaillard
- Department of Medical Oncology, European Hospital Georges Pompidou - APHP, Paris 75015, France
| | - Audrey Simonaggio
- Department of Medical Oncology, European Hospital Georges Pompidou - APHP, Paris 75015, France
| | - Stéphane Oudard
- Department of Medical Oncology, European Hospital Georges Pompidou - APHP, Paris 75015, France.,INSERM UMR-S1147, Paris 75006, France
| | - Yann-Alexandre Vano
- Centre de Recherche des Cordeliers, INSERM, Université de Paris, Sorbonne Université, Paris F-75006, France.,Department of Medical Oncology, European Hospital Georges Pompidou - APHP, Paris 75015, France
| |
Collapse
|
140
|
Corty RW, Langworthy BW, Fine JP, Buse JB, Sanoff HK, Lund JL. Antibacterial Use Is Associated with an Increased Risk of Hematologic and Gastrointestinal Adverse Events in Patients Treated with Gemcitabine for Stage IV Pancreatic Cancer. Oncologist 2020; 25:579-584. [PMID: 32181968 PMCID: PMC7356778 DOI: 10.1634/theoncologist.2019-0570] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 02/05/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Preclinical evidence has demonstrated that common intratumor bacteria metabolize the chemotherapeutic drug gemcitabine. The significance of this bacterial metabolism pathway, relative to the known metabolic pathways by host enzymes, is not known. We hypothesized that bacterial metabolism is clinically significant and that "knockdown" by antibacterial therapy has the unintended effect of increasing the effective dose of gemcitabine, thereby increasing the risk for gemcitabine-associated toxicities. MATERIALS AND METHODS We reanalyzed the comparator arm of the MPACT trial (NCT01442974), made available through Project Data Sphere, LLC (CEO Roundtable on Cancer's Life Sciences Consortium, Cary, NC; www.projectdatasphere.org). In this arm, 430 patients with metastatic pancreatic adenocarcinoma were treated with gemcitabine. We used the Anderson-Gill survival model to compare the risk of developing an adverse event after antibacterial prescription with time unexposed to antibacterials. Adverse events of grade 3 and greater were considered at three levels of granularity: all aggregated into one endpoint, aggregated by class, and taken individually. Antibiotic exposures were analyzed in aggregate as well as by class. RESULTS Antibacterial exposure was associated with an increased risk of adverse events (hazard ratio [HR]: 1.77; confidence interval [CI]: 1.46-2.14), any hematologic adverse event (HR: 1.64; CI: 1.26-2.13), and any gastrointestinal adverse event (HR: 2.14; CI: 1.12-4.10) but not a constitutional (HR: 1.33; CI: 0.611-2.90) or hepatologic adverse event (HR: 0.99; CI: 0.363-2.71). Among specific adverse events, antibacterial exposure was associated with an increased risk of anemia (HR: 3.16; CI: 1.59-6.27), thrombocytopenia (HR: 2.52; CI: 1.31-4.85), leukopenia (HR: 3.91; CI: 1.46-10.5), and neutropenia (HR: 1.53; CI: 1.07-2.17) but not any other specific adverse events. CONCLUSION Antibacterial exposure was associated with an increased risk of gemcitabine-associated, dose-limiting adverse events, including aggregate hematologic and gastrointestinal events, as well as four specific hematologic adverse events, suggesting that intratumor bacteria may be responsible for a clinically significant portion of gemcitabine metabolism. Alternative avenues of evidence will be necessary to confirm this preliminary finding and assess its generalizability. There is plentiful opportunity for similar analyses on other clinical trial data sets, where gemcitabine or other biomimetic small molecules were used. IMPLICATIONS FOR PRACTICE Patients treated with gemcitabine for metastatic pancreatic ductal adenocarcinoma have an increased rate of gemcitabine-associated toxicity during and after antibiotic therapy. This observation is consistent with preclinical evidence that intratumor bacteria metabolize gemcitabine to an inactive form. Further research is needed to determine whether this observation merits any changes in clinical practice.
Collapse
Affiliation(s)
- Robert W. Corty
- School of Medicine, University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Benjamin W. Langworthy
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Jason P. Fine
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - John B. Buse
- Translational and Clinical Sciences Institute, University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Hanna K. Sanoff
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Jennifer L. Lund
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| |
Collapse
|
141
|
Kapoor A, Noronha V, Patil VM, Joshi A, Menon N, Mahajan A, Janu A, Prabhash K. Concomitant use of antibiotics and immune checkpoint inhibitors in patients with solid neoplasms: retrospective data from real-world settings. Ecancermedicalscience 2020; 14:1038. [PMID: 32565891 PMCID: PMC7289608 DOI: 10.3332/ecancer.2020.1038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Indexed: 02/05/2023] Open
Abstract
Background The use of antibiotics is known to alter the gut microbiome and it is hypothesised that the use of antibiotics may also alter the response to immune checkpoint inhibitors (ICI). As data is limited from real-world settings, we performed a retrospective audit of patients who received ICI along with concomitant antibiotics. Patients and Methods This study is a retrospective audit of a prospectively collected the database of patients who received ICI for advanced solid tumours in any line between August 2015 and November 2018 at Tata Memorial Hospital, Mumbai, India. Antibiotic use was recorded from 2 weeks before the start of ICI and concomitantly with ICI. All statistical calculations were performed using Statistical Package for the Social Sciences (SPSS) statistical software for windows version 20.0. Results A total of 155 patients were identified as having received ICI during the study period, out of which 70 (44%) patients received antibiotics. Median PFS in patients who received antibiotics was 1.7 months (95% CI: 1.1–2.3) as against 3.6 months (95% CI: 2.3–4.8) for patients who did not receive antibiotics (p = 0.912). Median OS in the patients who received antibiotics was 3.9 months (95% CI: 1.8–11.4) as compared to 9.2 months (95% CI: 4.2–12.3) who did not receive antibiotics p = 0.053 (HR = 1.023; 95% CI: 1.00–1.04). Among the patients who received antibiotics, median OS for patients who received ≤10 days of antibiotics was 8.8 months (95% CI: 4.2–11.2) while for patients receiving >10 days of antibiotics, it was 2.8 months (95% CI: 1.2–4.4), p = 0.025 (HR = 2.0, 95% CI: 1.1–3.7). Thirty-three (21.2% of total) patients received antibiotics during the window of 2 weeks before the start of ICI to 2 months of starting ICI. Median OS in the patients who received antibiotics in this window was 2.8 months (95% CI: 1.2–4.5) as compared to 9.2 months (95% CI: 5.2–13.1) who did not receive antibiotics p = 0.008 (HR = 1.8; 95%CI: 1.2–3.0). Conclusions This study shows that the judicious use of antibiotics is required in patients on ICI or scheduled to be started on ICI.
Collapse
Affiliation(s)
- Akhil Kapoor
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, 400012, India.,http://orcid.org/0000-0001-6006-2631
| | - Vanita Noronha
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, 400012, India
| | - Vijay M Patil
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, 400012, India
| | - Amit Joshi
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, 400012, India
| | - Nandini Menon
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, 400012, India
| | - Abhishek Mahajan
- Department of Radiodiagnosis, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, 400012, India
| | - Amit Janu
- Department of Radiodiagnosis, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, 400012, India
| | - Kumar Prabhash
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, 400012, India
| |
Collapse
|
142
|
Derosa L, Routy B, Fidelle M, Iebba V, Alla L, Pasolli E, Segata N, Desnoyer A, Pietrantonio F, Ferrere G, Fahrner JE, Le Chatellier E, Pons N, Galleron N, Roume H, Duong CPM, Mondragón L, Iribarren K, Bonvalet M, Terrisse S, Rauber C, Goubet AG, Daillère R, Lemaitre F, Reni A, Casu B, Alou MT, Alves Costa Silva C, Raoult D, Fizazi K, Escudier B, Kroemer G, Albiges L, Zitvogel L. Gut Bacteria Composition Drives Primary Resistance to Cancer Immunotherapy in Renal Cell Carcinoma Patients. Eur Urol 2020; 78:195-206. [PMID: 32376136 DOI: 10.1016/j.eururo.2020.04.044] [Citation(s) in RCA: 202] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 04/20/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND The development of immune checkpoint blockade (ICB) has revolutionized the clinical outcome of renal cell carcinoma (RCC). Nevertheless, improvement of durability and prediction of responses remain unmet medical needs. While it has been recognized that antibiotics (ATBs) decrease the clinical activity of ICB across various malignancies, little is known about the direct impact of distinct intestinal nonpathogenic bacteria (commensals) on therapeutic outcomes of ICB in RCC. OBJECTIVE To evaluate the predictive value of stool bacteria composition for ICB efficacy in a cohort of advanced RCC patients. DESIGN, SETTING, AND PARTICIPANTS We prospectively collected fecal samples from 69 advanced RCC patients treated with nivolumab and enrolled in the GETUG-AFU 26 NIVOREN microbiota translational substudy phase 2 trial (NCT03013335) at Gustave Roussy. We recorded patient characteristics including ATB use, prior systemic therapies, and response criteria. We analyzed 2994 samples of feces from healthy volunteers (HVs). In parallel, preclinical studies performed in RCC-bearing mice that received fecal transplant (FMT) from RCC patients resistant to ICB (NR-FMT) allowed us to draw a cause-effect relationship between gut bacteria composition and clinical outcomes for ICB. The influence of tyrosine kinase inhibitors (TKIs) taken before starting nivolumab on the microbiota composition has also been assessed. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Metagenomic data (MG) from whole genome sequencing (WGS) were analyzed by multivariate and pairwise comparisons/fold ratio to identify bacterial fingerprints related to ATB or prior TKI exposure and patients' therapeutic response (overall response and progression-free survival), and compared with the data from cancer-free donors. RESULTS AND LIMITATIONS Recent ATB use (n = 11; 16%) reduced objective response rates (from 28% to 9%, p < 0.03) and markedly affected the composition of the microbiota, facilitating the dominance of distinct species such as Clostridium hathewayi, which were also preferentially over-represented in stools from RCC patients compared with HVs. Importantly, TKIs taken prior to nivolumab had implications in shifting the microbiota composition. To establish a cause-effect relationship between gut bacteria composition and ICB efficacy, NR-FMT mice were successfully compensated with either FMT from responding RCC patients or beneficial commensals identified by WGS-MG (Akkermansia muciniphila and Bacteroides salyersiae). CONCLUSIONS The composition of the microbiota is influenced by TKIs and ATBs, and impacts the success of immunotherapy. Future studies will help sharpen the role of these specific bacteria and their potential as new biomarkers. PATIENT SUMMARY We used quantitative shotgun DNA sequencing of fecal microbes as well as preclinical models of fecal or bacterial transfer to study the association between stool composition and (pre)clinical outcome to immune checkpoint blockade. Novel insights into the pathophysiological relevance of intestinal dysbiosis in the prognosis of kidney cancer may lead to innovative therapeutic solutions, such as supplementation with probiotics to prevent primary resistance to therapy.
Collapse
Affiliation(s)
- Lisa Derosa
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; Faculté de Médecine Kremlin-Bicêtre, Université Paris Sud, Université Paris Saclay, France; Department of Medical Oncology, Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
| | - Bertrand Routy
- Hematology-Oncology Division, Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada; Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal l, Canada CHUM, Montréal, QC, Canada
| | - Marine Fidelle
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; Faculté de Médecine Kremlin-Bicêtre, Université Paris Sud, Université Paris Saclay, France; Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France; Faculté de Pharmacie, University Paris-Saclay, Chatenay-Malabry, France
| | - Valerio Iebba
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Laurie Alla
- Université Paris-Saclay, INRAE MetaGenoPolis, Jouy-en-Josas, France
| | - Edoardo Pasolli
- Department of Agricultural Sciences, University of Naples Federico II, Napoli, Italy
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy; Istituto Europeo di Oncologie, Milan, Italy
| | - Aude Desnoyer
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Faculté de Pharmacie, University Paris-Saclay, Chatenay-Malabry, France; Gustave Roussy Cancer Campus, Laboratory of Immunomonitoring in Oncology, CNRS-UMS 3655 and INSERM-US23, Villejuif, France
| | | | - Gladys Ferrere
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Jean-Eudes Fahrner
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; Faculté de Médecine Kremlin-Bicêtre, Université Paris Sud, Université Paris Saclay, France; Transgene S.A., Illkirch-Graffenstaden, France
| | | | - Nicolas Pons
- Université Paris-Saclay, INRAE MetaGenoPolis, Jouy-en-Josas, France
| | | | - Hugo Roume
- Université Paris-Saclay, INRAE MetaGenoPolis, Jouy-en-Josas, France
| | - Connie P M Duong
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Laura Mondragón
- Faculté de Médecine Kremlin-Bicêtre, Université Paris Sud, Université Paris Saclay, France; Cell Biology and Metabolomics Platforms, Gustave Roussy Cancer Campus, Villejuif, France; Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
| | | | - Mélodie Bonvalet
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Safae Terrisse
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; Faculté de Médecine Kremlin-Bicêtre, Université Paris Sud, Université Paris Saclay, France; Department of Medical Oncology, Gustave Roussy Cancer Campus (GRCC), Villejuif, France
| | - Conrad Rauber
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; Faculté de Médecine Kremlin-Bicêtre, Université Paris Sud, Université Paris Saclay, France
| | - Anne-Gaëlle Goubet
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; Faculté de Médecine Kremlin-Bicêtre, Université Paris Sud, Université Paris Saclay, France
| | | | | | - Anna Reni
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Beatrice Casu
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Maryam Tidjani Alou
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Carolina Alves Costa Silva
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Didier Raoult
- Aix-Marseille Université, MEPHI, IRD, IHU Méditerranée Infection, Marseille, France
| | - Karim Fizazi
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Department of Medical Oncology, Gustave Roussy Cancer Campus (GRCC), Villejuif, France
| | - Bernard Escudier
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Department of Medical Oncology, Gustave Roussy Cancer Campus (GRCC), Villejuif, France
| | - Guido Kroemer
- Cell Biology and Metabolomics Platforms, Gustave Roussy Cancer Campus, Villejuif, France; Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France; EverImmune, GRCC, Villejuif, France; Aix-Marseille Université, MEPHI, IRD, IHU Méditerranée Infection, Marseille, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China; Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Laurence Albiges
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Faculté de Médecine Kremlin-Bicêtre, Université Paris Sud, Université Paris Saclay, France; Department of Medical Oncology, Gustave Roussy Cancer Campus (GRCC), Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; Faculté de Médecine Kremlin-Bicêtre, Université Paris Sud, Université Paris Saclay, France; Department of Medical Oncology, Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France; EverImmune, GRCC, Villejuif, France; Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China.
| |
Collapse
|
143
|
Microbial Alterations and Risk Factors of Breast Cancer: Connections and Mechanistic Insights. Cells 2020; 9:cells9051091. [PMID: 32354130 PMCID: PMC7290701 DOI: 10.3390/cells9051091] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 12/12/2022] Open
Abstract
Breast cancer-related mortality remains high worldwide, despite tremendous advances in diagnostics and therapeutics; hence, the quest for better strategies for disease management, as well as the identification of modifiable risk factors, continues. With recent leaps in genomic technologies, microbiota have emerged as major players in most cancers, including breast cancer. Interestingly, microbial alterations have been observed with some of the established risk factors of breast cancer, such as obesity, aging and periodontal disease. Higher levels of estrogen, a risk factor for breast cancer that cross-talks with other risk factors such as alcohol intake, obesity, parity, breastfeeding, early menarche and late menopause, are also modulated by microbial dysbiosis. In this review, we discuss the association between known breast cancer risk factors and altered microbiota. An important question related to microbial dysbiosis and cancer is the underlying mechanisms by which alterations in microbiota can support cancer progression. To this end, we review the involvement of microbial metabolites as effector molecules, the modulation of the metabolism of xenobiotics, the induction of systemic immune modulation, and altered responses to therapy owing to microbial dysbiosis. Given the association of breast cancer risk factors with microbial dysbiosis and the multitude of mechanisms altered by dysbiotic microbiota, an impaired microbiome is, in itself, an important risk factor.
Collapse
|
144
|
Oncobiosis and Microbial Metabolite Signaling in Pancreatic Adenocarcinoma. Cancers (Basel) 2020; 12:cancers12051068. [PMID: 32344895 PMCID: PMC7281526 DOI: 10.3390/cancers12051068] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic adenocarcinoma is one of the most lethal cancers in both men and women, with a median five-year survival of around 5%. Therefore, pancreatic adenocarcinoma represents an unmet medical need. Neoplastic diseases, such as pancreatic adenocarcinoma, often are associated with microbiome dysbiosis, termed oncobiosis. In pancreatic adenocarcinoma, the oral, duodenal, ductal, and fecal microbiome become dysbiotic. Furthermore, the pancreas frequently becomes colonized (by Helicobacter pylori and Malassezia, among others). The oncobiomes from long- and short-term survivors of pancreatic adenocarcinoma are different and transplantation of the microbiome from long-term survivors into animal models of pancreatic adenocarcinoma prolongs survival. The oncobiome in pancreatic adenocarcinoma modulates the inflammatory processes that drive carcinogenesis. In this review, we point out that bacterial metabolites (short chain fatty acids, secondary bile acids, polyamines, indole-derivatives, etc.) also have a role in the microbiome-driven pathogenesis of pancreatic adenocarcinoma. Finally, we show that bacterial metabolism and the bacterial metabolome is largely dysregulated in pancreatic adenocarcinoma. The pathogenic role of additional metabolites and metabolic pathways will be identified in the near future, widening the scope of this therapeutically and diagnostically exploitable pathogenic pathway in pancreatic adenocarcinoma.
Collapse
|
145
|
Moving towards personalized treatments of immune-related adverse events. Nat Rev Clin Oncol 2020; 17:504-515. [DOI: 10.1038/s41571-020-0352-8] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2020] [Indexed: 12/17/2022]
|
146
|
Schnell A, Bod L, Madi A, Kuchroo VK. The yin and yang of co-inhibitory receptors: toward anti-tumor immunity without autoimmunity. Cell Res 2020; 30:285-299. [PMID: 31974523 PMCID: PMC7118128 DOI: 10.1038/s41422-020-0277-x] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 01/06/2020] [Indexed: 12/31/2022] Open
Abstract
Co-inhibitory receptors are important regulators of T-cell function that define the balance between tolerance and autoimmunity. The immune regulatory function of co-inhibitory receptors, including CTLA-4, PD-1, TIM-3, TIGIT, and LAG-3, was first discovered in the setting of autoimmune disease models, in which their blockade or deficiency resulted in induction or exacerbation of the disease. Later on, co-inhibitory receptors on lymphocytes have also been found to influence outcomes in tumor and chronic viral infection settings. These receptors suppress T-cell function in the tumor microenvironment (TME), thereby making the T cells dysfunctional. Based on this observation, blockade of co-inhibitory receptors (also known as checkpoint molecules) has emerged as a successful treatment option for a number of human cancers. However, severe autoimmune-like side effects limit the use of therapeutics that block individual or combinations of co-inhibitory receptors for cancer treatment. In this review we provide an overview of the role of co-inhibitory receptors in autoimmunity and anti-tumor immunity. We then discuss current approaches and future directions to leverage our knowledge of co-inhibitory receptors to target them in tumor immunity without inducing autoimmunity.
Collapse
Affiliation(s)
- Alexandra Schnell
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Lloyd Bod
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Asaf Madi
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| |
Collapse
|
147
|
NSCLC Immunotherapy Efficacy and Antibiotic Use: A Systematic Review and Meta-Analysis. J Thorac Oncol 2020; 15:1147-1159. [PMID: 32173463 DOI: 10.1016/j.jtho.2020.03.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/17/2020] [Accepted: 03/01/2020] [Indexed: 02/07/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically improved patient outcomes in a variety of tumor types, but with variable efficacy. Recent research has suggested that antibiotic-induced disruption of the microbiota may impact ICI efficacy. We performed a systematic review and meta-analysis of studies that assessed the impact of antibiotic use on the survival of patients diagnosed with NSCLC and treated with ICI. We systematically searched Medline, the Cochrane Library, and major oncology conferences proceedings. Eligible studies mentioned hazard ratio or Kaplan-Meier curves for progression-free survival (PFS) or overall survival (OS) based on antibiotic exposure before or during ICI treatment. We identified 23 eligible studies. The impact of antibiotics was then evaluated in 2208 patients for PFS and 5560 for OS. For both PFS and OS meta-analyses, the between-study heterogeneity was high (Higgins and Thompson I2 of 69% and 80%, respectively). The pooled hazard ratio was 1.47 (95% confidence interval [CI]: 1.13-1.90) for PFS and 1.69 (95% CI: 1.25-2.29) for OS revealing a significantly reduced survival in patients with NSCLC exposed to antibiotics. The median OS was reduced on average by 6.7 months (95% CI: 5.1-8.4) in the patients exposed to antibiotics. The effect seems to depend on the time window of exposure with stronger effects reported when the patients took antibiotics [-60 days; +60 days] around ICI initiation. In patients with NSCLC, the findings of the meta-analysis indicate that antibiotic use before or during treatment with ICI leads to a median OS decreased by more than 6 months. Specifically, exposure shortly before or after ICI initiation seems to be particularly detrimental, whereas antibiotic use later during disease course does not seem to alter survival. Because PFS and OS were difficult to compare between studies owing to heterogeneity and the multiple confounding factors identified, further studies are needed to strengthen the understanding of this phenomenon.
Collapse
|
148
|
Casadei C, Lolli C, Farolfi A. Immune-checkpoint inhibitors and the importance of concomitant medications: focus on antibiotics. ANNALS OF TRANSLATIONAL MEDICINE 2020; 7:S339. [PMID: 32016057 DOI: 10.21037/atm.2019.09.99] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Chiara Casadei
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Italy
| | - Cristian Lolli
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Italy
| | - Alberto Farolfi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Italy
| |
Collapse
|
149
|
Elkrief A, Joubert P, Florescu M, Tehfe M, Blais N, Routy B. Therapeutic landscape of metastatic non-small-cell lung cancer in Canada in 2020. ACTA ACUST UNITED AC 2020; 27:52-60. [PMID: 32218661 DOI: 10.3747/co.27.5953] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lung cancer is the most commonly diagnosed cancer in Canada and remains associated with high mortality. Nevertheless, recent advances in the fields of immuno-oncology and precision medicine have led to significant improvements in clinical outcome in metastatic non-small-cell lung cancer (nsclc). Those improvements were facilitated by a greater understanding of the biologic classification of nsclc, which catalyzed discoveries of novel therapies. Here, we present a comprehensive review of the recent avalanche of practice-changing trials in metastatic nsclc, and we offer an approach to the management of this disease from a Canadian perspective. We begin with an overview of the pathologic and molecular characterization of metastatic nsclc. Next, we review the indications for currently approved immune checkpoint inhibitors, and we provide an approach to the management of disease with a driver mutation. Finally, we address future avenues in both diagnostics and therapeutics for patients with advanced and metastatic nsclc.
Collapse
Affiliation(s)
- A Elkrief
- Segal Cancer Centre, Sir Mortimer B. Davis Jewish General Hospital, Montreal.,Cedars Cancer Centre, McGill University Health Centre, Montreal.,Centre de recherche du Centre hospitalier de l'Universtité de Montréal, Montreal
| | - P Joubert
- Institut universitaire de cardiologie et de pneumologie de Québec, Quebec City
| | - M Florescu
- Centre de recherche du Centre hospitalier de l'Universtité de Montréal, Montreal.,Division of Hematology-Oncology, Centre hospitalier de l'Université de Montréal, Montreal, QC
| | - M Tehfe
- Centre de recherche du Centre hospitalier de l'Universtité de Montréal, Montreal.,Division of Hematology-Oncology, Centre hospitalier de l'Université de Montréal, Montreal, QC
| | - N Blais
- Centre de recherche du Centre hospitalier de l'Universtité de Montréal, Montreal.,Division of Hematology-Oncology, Centre hospitalier de l'Université de Montréal, Montreal, QC
| | - B Routy
- Centre de recherche du Centre hospitalier de l'Universtité de Montréal, Montreal.,Division of Hematology-Oncology, Centre hospitalier de l'Université de Montréal, Montreal, QC
| |
Collapse
|
150
|
Abdel-Rahman O, Ghosh S, Walker J. Outcomes of metastatic colorectal cancer patients in relationship to prior and concurrent antibiotics use; individual patient data analysis of three clinical trials. Clin Transl Oncol 2020; 22:1651-1656. [PMID: 32008218 DOI: 10.1007/s12094-020-02301-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 01/09/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND Antibiotic use at the time of chemotherapy has been linked with inferior outcomes among a number of solid tumors. The current study aims at further assessing this observation among metastatic colorectal cancer patients treated with first-line systemic chemotherapy. METHODS This is a pooled analysis of three clinical trial datasets (NCT00384176; NCT00272051; NCT00305188) that were accessed from the Project Data Sphere platform. Kaplan-Meier survival estimates were used to evaluate the impact of antibiotic use on overall and progression-free survival and multivariable Cox regression models were employed to further assess this impact. RESULTS A total of 1446 patients were included in the current analysis. These include 108 patients who received antibiotics before the start of chemotherapy, 499 patients who received antibiotics after the start of chemotherapy, and 839 patients who did not receive antibiotics. Using Kaplan-Meier survival estimates, the use of antibiotics prior to the start of chemotherapy was associated with worse progression-free (P = 0.001) and overall survival (P < 0.001). Likewise, when multivariable Cox regression analyses were conducted, prior antibiotic use is associated with worse progression-free (HR for antibiotic use during chemotherapy versus antibiotic use prior to chemotherapy = 0.764; 95% CI 0.604-0.966; P = 0.024) and overall survival (HR for antibiotic use during chemotherapy versus antibiotic use prior to chemotherapy = 0.710; 95% CI 0.537-0.940; P = 0.017). CONCLUSION Antibiotic use before (but not following) the start of 5FU-based chemotherapy is associated with worse progression-free and overall survival among patients with metastatic colorectal cancer.
Collapse
Affiliation(s)
- O Abdel-Rahman
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, T4G1Z2, Canada.
| | - S Ghosh
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, T4G1Z2, Canada
| | - J Walker
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, T4G1Z2, Canada
| |
Collapse
|