Skip to main content

MINI REVIEW article

Front. Oncol., 24 September 2020
Sec. Thoracic Oncology
This article is part of the Research Topic Impact of Immunotherapy in Lung Cancer View all 9 articles

Microbiota and Lung Cancer. Opportunities and Challenges for Improving Immunotherapy Efficacy

\nMaitane Ocriz-Díez,
Maitane Ocáriz-Díez1,2*Mara Cruellas,Mara Cruellas1,2Marta Gascn,Marta Gascón1,2Rodrigo Lastra,Rodrigo Lastra1,2Luis Martínez-Lostao,,,,Luis Martínez-Lostao2,3,4,5,6Ariel Ramírez-LabradaAriel Ramírez-Labrada7Jos Ramn Pao,José Ramón Paño2,8Andrea Sesma,Andrea Sesma1,2Irene Torres,Irene Torres1,2Alfonso Yubero,Alfonso Yubero1,2Julin Pardo,,,Julián Pardo2,9,10,11Dolores Isla,Dolores Isla1,2Eva M. GlvezEva M. Gálvez12
  • 1Medical Oncology Department, Lozano Blesa University Clinical Hospital, Zaragoza, Spain
  • 2Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
  • 3Inmunology Department, Lozano Blesa University Clinical Hospital, Zaragoza, Spain
  • 4Department of Microbiology, Pediatrics, Radiology and Public Health, University of Zaragoza, Zaragoza, Spain
  • 5Aragon Nanoscience Institute, Zaragoza, Spain
  • 6Aragon Materials Science Institute, Zaragoza, Spain
  • 7Unidad de Nanotoxicología e Inmunotoxicología (UNATI), Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
  • 8Infectious Diseases Department, Lozano Blesa University Clinical Hospital, Zaragoza, Spain
  • 9ARAID Foundation (IIS Aragón), Zaragoza, Spain
  • 10Microbiology, Preventive Medicine and Public Health Department, Medicine, University of Zaragoza, Zaragoza, Spain
  • 11Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine Network (CIBER-BBN), Madrid, Spain
  • 12Instituto de Carboquimica (ICB-Consejo Superior de Investigaciones Cientificas), Zaragoza, Spain

The advances in molecular biology and the emergence of Next Generation Sequencing (NGS) have revealed that microbiome composition is closely related with health and disease, including cancer. This relationship affects different levels of cancer such as development, progression, and response to treatment including immunotherapy. The efficacy of immune checkpoint inhibitors (ICIs) may be influenced by the concomitant use of antibiotics before, during or shortly after treatment with ICIs. Nevertheless, the linking mechanism between microbiote, host immunity and cancer is not clear and the role of microbiota manipulation and analyses in cancer management has not been clinically validated yet. Regarding the use of microbiome as biomarker to predict ICI efficacy it has been recently shown that the use of biochemical serum markers to monitor intestinal permeability and loss of barrier integrity, like citrulline, could be useful to monitor microbiota changes and predict ICI efficacy. There are still many unknowns about the role of these components, their relationship with the microbiota, with the use of antibiotics and the response to immunotherapy. The next challenge in microbiome research will be to identify individual microbial species that causally affect lung cancer phenotypes and response to ICI and disentangle the underlying mechanisms. Thus, further analyses in patients with lung cancer receiving treatment with ICIs and its correlation with the composition of the microbiota in different organs including the respiratory tract, peripheral blood and intestinal tract could be useful to predict the efficacy of ICIs and its modulation with antibiotic use.

Introduction

Lung cancer is the most common cancer and the most frequent cause of cancer-related deaths worldwide both in men and women (1). Every year, more than 1.8 million people are diagnosed with lung cancer, accounting for 1.6 million deaths as a result of the disease. 5-year survival rates vary between 4 and 17% depending on stage and national differences (2). Non-small cell lung cancer (NSCLC) accounts for 80%−90% of lung cancers, while small cell lung cancer (SCLC) has been decreasing in frequency in many countries over the past two decades (1). Tobacco consumption represents one of the main risk factors for lung cancer due to the presence of carcinogens that promote cell transformation. However, other factors like chronic inflammatory immune-mediated responses, genetic predisposition or infections, like viral or more recently described bacterial, have been involved in lung carcinogenesis (3).

The clinical management of this neoplasm has undergone constant modifications, due to better knowledge of the biology of the tumor and the genomic profile of each patient, which allows a more accurate diagnosis and efficient treatment. During the last few years, new therapies directed against molecular targets involved in the regulation of the immune homeostasis and cancer immunity have been incorporated, particularly immune checkpoint inhibitors (ICIs), such as monoclonal antibodies against the Programmed Cell Death Protein 1/Ligand 1 (PD1/L1) axes or Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4) (4). These molecules are critical negative regulators of the immune system that in physiological conditions prevent reactions against self-antigens avoiding autoimmune reactions. Some tumors have learnt to use these molecules to avoid immune attack and perpetuate in the host, and thus, under specific situations blocking ICs release these brakes and promote immune-mediated tumor elimination.

ICIs are effective in locally advanced unresectable NSCLC as consolidation after chemoradiotherapy (durvalumab) (5), in patients with pretreated metastatic disease (nivolumab, pembrolizumab, atezolizumab) (69) and also as first-line treatment, both as monotherapy (pembrolizumab) (10), in combination with chemotherapy (pembrolizumab, atezolizumab) (11, 12) or as double immunotherapy (anti-PD1 nivolumab/anti-CTLA4 ipilimumab) (13). Recently it has also been found to improve outcomes in metastatic SCLC as first line treatment in combination with chemotherapy (atezolizumab, durvalumab) (14, 15).

Despite these positive results, the major challenge that clinicians face is the identification of those patients that will benefit most from this therapy, including efficacy and low toxicity. At present, the most commonly used biomarker to predict ICIs efficacy, PDL1 expression, presents several limitations (16).

In addition to prevent unwanted reactions against self-antigens, ICIs are key molecules involved in the homeostatic regulation of the immune response against commensal microbiota. Thus, in recent years, microbiota has emerged as critical regulator of the efficacy and toxicity of ICIs in different types of cancer including lung. Here after a brief summary of the main experimental evidences relating microbiota with lung cancer development, we will discuss the relationship between the composition of lung microbiota and the efficacy of ICIs and the opportunities that this connection offers to exploit microbiome analyses and modification in the prediction of ICIs efficacy (17). Finally, we will present some of the main challenges that will be required to overcome in order to use microbiota modification as a therapeutic approach to enhance ICIs efficacy and safety.

Microbiota and Tissue Homeostasis

The microbiota is emerging as a key regulator of cancer development and treatment. From birth and throughout whole life, the human beings keep an intimate mutually beneficial relationship with the microbial community, using it as protection against external aggressions and as a supplier of beneficiary essential molecules.

The human microbiota is the ecological community of commensal, symbiotic and pathogenic microorganisms, including protozoa, fungi, bacteria, and viruses. Microbiota is acquired after birth by vertical transmission inheritance from the mother through direct surface contact, beginning shortly after that its evolution (in response to environmental factors, diet, drugs or environmental exposures) cumulating in a stable adult microbiota by the age of two (18). Most studies have been focused on the bacterial composition of microbiota, since these microorganisms are the most abundant within commensal microbiota. Indeed, it is estimated that a single human subject harbors more than one billion bacterial cells, most of which are commensal, so the total microbiota is estimated to constitute 0.2 kg of weight (19).

The size and the composition of the microbiota differ by anatomical site, showing a wide interpersonal variation but relatively temporal stability in a single individual (20). Thus, the individual composition of microbiota should be taken into account when using it as a biomarker for cancer treatment as well as to design effective microbiota transplant protocols, which are major current challenges in different pathologies including lung cancer, as will be discussed in the next sections.

As indicated above, among other functions, microbiota regulates host immunity and tissue homeostasis. Therefore, the same microorganisms that are beneficial to human health, under certain circumstances, can promote the development of diseases and cancer (2125). The eubiosis is conditioned by multiple factors: hereditary and environmental factors, genetic background, diseases, lifestyle (mainly diet), chronic infections or antibiotic exposure, among others (26). These factors can contribute to the perturbation of the balance of microbiota composition, a situation known as dysbiosis. This situation can be mild and temporal reverting after the detrimental stimulus is removed. However, in some situations, this dysbalance can be chronified, altering tissue homeostasis and leading to diseases like cancer (25). Direct effects of microbial components on cell transformation or indirect effects related with a dysregulated inflammatory immune response have been found to be involved in carcinogenesis (27).

Lung Microbiota, Cancer and Immunotherapy

The gut microbiota gives the major contribution to human microbiota and comprises about 3 × 1013 bacterial cells, followed by the skin, which is estimated to harbor ~1012 bacteria (28). Accordingly, the intestinal microbiota and its role in regulating host metabolism and gastrointestinal and metabolic diseases such as gastric cancer (29, 30), inflammatory bowel disease (a risk factor for colorectal cancer), diabetes, and obesity (3133) is the most studied so far. However, recent findings indicate that other microbial populations in the body like lungs are very important in keeping tissue homeostasis and contribute to diseases like asthma, COPD or lung cancer (27).

Lungs have traditionally been considered sterile space, nevertheless they are constantly exposed to microorganisms since the lower respiratory tract is replete with diverse communities of bacteria both in health and in diseased states (34). Bacterial communities in the lungs are dynamic as the airways are constantly exposed to microorganisms suspended in the air that flow through the oral cavity and upper respiratory tract to the lower respiratory tract. The composition of lung microbiota modulates a healthy host lung immune system. A proper balance between lung immunity and microbiota is key to fight infection but, in addition, to prevent lung inflammatory diseases like cancer. The epithelial cell barrier is the first line of defense in lungs preventing invasion by pathogenic microbes and, at the same time, establishing key molecular interactions with the commensal microbiota that regulates immune homeostasis. Commensal microbiota regulates immune tolerance, decreasing lung inflammation through dendritic cell (DC), γδ T, and T regulatory (Treg) cell recruitment (27). Other resident immune cells like macrophages, Innate Lymphoid Cells (ILCs) and T cell subsets contribute to regulate local inflammatory responses by preventing the overload of pathogens or their metabolites. Lung microbiota dysbiosis can contribute to cancer development by inducing a dysregulated immune response characterized by an overactivation of inflammatory cells like M1 macrophages, Th1 cells or γδ T cells. Inflammation contributes to cell transformation and cancer by activating cell proliferation pathways as well as by inducing radical oxygen species and genetic mutations. The specific connection between airway microbiota, immune regulation and lung cancer is not clear yet. As an example it was recently shown that the presence of a lung bacterial composition enriched in oral commensals is associated with a prevalence of T helper 17 (Th17) inflammation that seems to be key for a proper regulation of the lung immune response (35). However, the connection between Th17 cells, chronic lung inflammation and cancer is not yet clear.

Although less explored than the connection between microbiota dysbiosis, chronic inflammation and cancer development, microbial components can contribute to lung cancer development by producing metabolites with oncogenic potential. For example, Tsay et al. (36) found that exposure to Veillonella, Prevotella, and Streptococcus bacteria induce epithelial cell transformation in vitro and in vivo by activating the PI3K and ERK pathways.

The respiratory microbiome differs significantly between the three major anatomical regions: oral/nasal, upper and lower respiratory tract (37). Taking into account the communication between the oral cavity and the respiratory tract, the oral microbiome could be highly relevant to the lung microbiome. Nevertheless, not every alteration observed in sputum represents changes in the microbiome of the lower airway microbiome, since there is increasing evidence that the microbiome of sputum is a better reflection of the oral microbiome than of the lower airway microbiome (3840).

Movement of microbes between these sites and the lungs occurs regularly via breathing and microaspiration of pharyngeal secretions, which seems to be the main source of lung microbiota in healthy subjects (41). Despite this connection, it should be considered that the composition and function of upper and lower airways microbiome are not identical and (37), thus, these differences should be had in mind to design studies that can be successfully applied to the diagnosis and treatment of cancer. For example, identification of microbiome composition in oral cavity or saliva could be useful to identify biomarkers to predict ICI response, but when designing interventions to modulate lung microbiota composition and enhance ICIs efficacy, genuine identification of lung microbiota might be required.

Several sample types are used to analyzed lung microbiome composition: bronchoalveolar lavage (BAL), bronchial brushing tissue, buccal sample, surgical resection tissue or exhaled breath condensate (23, 42), among others. Large number of samples, use of non-invasive studies, and exact statistical analysis are crucial for detection of predominant bacterial species in lung cancer (43). Because of the mentioned technical reasons, characterization of the oral microbiome is less complex than characterization of lung microbiome. The oral microbiota includes bacteria, fungi, as well as viruses. It has been associated with systemic diseases including infective endocarditis, rheumatoid arthritis, pulmonary disease, and in more recent studies, with cancer (4446). It is important to take into account that the oral microbiota can aid in conversion of alcohol and smoking byproducts to mutagenic compounds, so that the oral dysbiosis may be related to premalignant processes and the cancer itself (34). Thus, analyses of oral microbiota might also be interesting to find out predictive biomarkers for ICI or even to modulate its composition to prevent cancer development or progression or to enhance ICI efficacy.

The number of studies in the field of lung microbiota is rapidly growing and have yielded provocative observations highlighting a possible relationship between lung microbiota and respiratory disease (23). The development of lung cancer seems to be closely associated with chronic inflammation characterized by infiltration of inflammatory cells and accumulation of pro-inflammatory factors (cytokines, chemokines, and prostaglandins), a high bacterial load and linked to airway dysbiosis or reduction in the dominant bacterial species in the microbiota (37, 4749).

Specific microbes have been associated with lung cancer (Pseudomona, Veillonella, Streptococcus, Provotella, Fusobacteria…) (50, 51) as Bacteroides, Firmicutes, and Proteobacteria are the most common phyla consistently observed in lung microbiota of healthy individuals (23, 52). Higher richness and diversity in normal tissue seems to be significantly associated with reduced recurrence-free and disease-free survival. Family Lachnospiraceae, genera Faecalibacterium and Ruminococcus, among others, are associated with reduced recurrence-free and disease-free survival (53).

Paramount to the development of microbiota-based therapeutics, the next challenge in microbiome research will be to identify individual microbial species that causally affect cancer phenotypes and unravel the underlying mechanisms (54). Metagenomics studies using culture-independent techniques such as PCR amplification and sequencing of the 16S ribosomal (r)RNA gene, especially variable regions V3-V4 of 16S rRNA bacterial gene sequencing, enables the identification and classification of the species without requiring pre-cultivation (25, 55). The sequencing of basal stool and saliva samples from patients treated with ICIs has obtained striking preliminary results that suggest a close relationship between the commensal microbiome and the clinical efficacy of immunotherapy, which might be useful to stratify patients based on their metagenomic microbial fingerprint (28). However, it should be taken into account that rRNA sequencing has limitations since it cannot differentiate between living and dead microbes. Thus, metagenomic analyses just provide the picture of the microbial composition (microbiome) without providing information on the potential functional consequences of those microbes. Here the analyses of genes expressed by the microbial community (metatranscriptomics) and their byproducts (metabolomics) can be very useful since they provide information on the functional state of microbiota and the potential consequences for the health of the environmental niche they inhabit, the lung in this case. Thus, the combination of metagenomics, metatranscriptomics, and metabolomics will provide a whole picture on the composition of lung microbiota and their functional consequences under different conditions (56).

ICIs act by blocking pathways of negative regulation of the immune system, in order to enhance antitumor immune response. Due to the general dysregulation of immune homeostasis, ICIs induce a broad spectrum of side effects potentially involving any organ, known as immune-related adverse events (irAEs) (57). irAEs occur more commonly in patients on anti-CTLA- 4 treatment as compared with those taking anti-PD-1/-L1, and the incidence of irAEs increases when these agents are used in combination. It has been reported that patients who receive ICIs and have immuno-related toxicity (58, 59) have a different intestinal microbiota composition, in addition to a possible greater benefit of treatment (60, 61). Thus, further studies correlating these factors are of interest in order to establish microbiome composition as a predictive biomarker for ICI efficacy and toxicity.

There are multiple studies exploring the possibility that dysbiosis associated with malignant disease or concomitant antibiotic use (before, during or shortly after treatment with ICIs) could influence ICI efficacy. NSCLC patients taking antibiotic during ICI treatment have a significant reduction in progression free survival and overall survival compared with those who did not receive antibiotics (62). Here it should be taken into consideration that antibiotic use might be associated with a more severe disease and thus, this might act as a confounding factor when studying the correlation between antibiotic and ICI efficacy. Although most studies have focused on the intestinal microbiota, it is expected that antibiotics could also influence the lung microbiota, altering the antitumoral effect of ICIs, anti-CTLA4 and anti-PD-1 / L1 in patients with lung cancer (63).

Mucosal Barrier Integrity and Microbiota

Gut and lung microbiota are connected thanks to a complex bidirectional axis via lymphatic and blood circulation, which may explain why changes in one mucosal compartment could directly impact a distant mucosal site (64).

Gut microbes can influence lung immune function through different mechanisms. One would be the connection through molecular patterns associated with pathogens, such as lipopolysaccharides (LPS), which can stimulate toll-like receptors and activate genes that regulate inflammation and responses of the innate immune system. In addition, they would cause phenotypic changes in dendritic cells (DCs) and migration to mesenteric lymphoid nodes to promote the priming of T lymphocytes and the production of various regulatory cytokines (IL-10, TGF-β, IFNγ, and IL-6). In these lymph nodes, T cells acquire localization molecules that initiate migration to the respiratory mucosa, through CCR4 or CCR6, where they promote protective and anti-inflammatory responses. Another mechanism would be metabolites such as short chain fatty acids (SCFA), produced by the fermentation of carbohydrates by bacteria, which modify gene expression through the inhibition of histone deacetylases, methylation, the production of cytokines and chemokines, and cell differentiation, proliferation and apoptosis (18, 6567).

Intestinal dysbiosis leads to perturbation of gut immune homeostasis favoring the development of inflammatory processes and even tumoral processes (30, 57). The permeability of the epithelial barriers, like intestinal and respiratory tract, is closely related to the luminal content and the interaction with the microbiota. Permeability depends on the junctional complex between the intestinal enterocytes, which includes tight junction (TJ) proteins that regulate the transport of ions and water between the gut lumen and the blood stream. Increased gut permeability causes the trespass of antigens to the lamina propia and the blood stream, promoting a local and systemic inflammatory immune responses enhancing epithelial barrier loss and affecting distant organs and tissues (68). The protein zonuline is an admissible regulator of TJs permeability and the only known physiological regulator of intestinal TJs, and its presence in serum has been proposed as a peripheral marker to assess gut permeability (69), as well as endotoxin. (70, 71). Thus, the loss of barrier function may be suspected by a positive zonuline regulation (68, 7274). However, there are many unknowns about the role of these components, their relationship with the microbiota, with the use of antibiotics and with the response to immunotherapy. In addition, it has been recently shown that detection of zonuline in serum might be affected by serious technical flaws and thus its value to monitor epithelial barrier permeability should be confirmed (75).

On the other hand, plasma citrulline, an amino acid produced by enterocytes, has been validated as a marker of the intestinal barrier and function of enterocytes. Citrulline can be produced as a result of a post-transduction modification from arginine in a reaction catalyzed by peptidyl-arginine deaminase. This modification occurs in inflammatory processes and cell death. In these processes, citrulline can be recognized aberrantly by the immune system, leading to the generation of anti-citrulline antibodies. Under some circumstances, such as infections and the need for antibiotic treatment in patients with lung cancer undergoing treatment with ICIs, inflammatory phenomena and cell death may occur leading to aberrant recognition of citrulline and the generation of anti-citrulline antibodies. Finally, it seems that citrulline levels are affected with the use of antibiotics and can be correlated with the response to immunotherapy (76). Identification of soluble circulating factors that might be markers of the integrity of the epithelial barrier could be very useful to monitor cancer progression and treatment efficacy as discussed below.

Several studies have studied the correlation between gut microbiome and outcomes to ICIs. A greater diversity of the intestinal microbiome seems to be related to a greater progression-free survival (77), showing a higher concentration of Akkermansia muciniphilia in the stools of the responders (62).

Challenges and Future Perspectives

Understanding how microorganisms present in the respiratory tract can influence the development of lung carcinoma and the effectiveness of treatments is key to develop therapeutic approaches to modify the microbial composition and thus improve their effectiveness.

Future precision medicine strategies will likely be based on new diagnostic and therapeutic tools that identify alterations in the microbiome in each patient that could be used as new biomarkers for individual treatments (28). These biomarkers could also help identify candidate patients for microbiota intervention strategies such as fecal microbiota transplantation (FMT) with the possibility of evaluating the effects of selected members of the commensal gut microbiota on systemic immunity, including in the lungs, as well as the use of probiotics and prebiotics (78, 79). FMT, usually used to treat recurrent Clostridium difficile infection, could be used as therapy to treat other diseases related to an unhealthy gut microbiota. However, before implementing these approaches it should be clarified how to prepare patients for FMT (preconditioning) and how to manage possible complications related to antibiotic resistance and the potential of infections due to FMT.

The modulation of microbiota is considered a relevant target for novel therapeutic and preventive treatments against a range of diseases (70), albeit this field is still in its infancy and more research is required before microbiota can be safely manipulated to prevent or treat disease. The microbiome may be potentially manipulated with an aim to correct dysbiosis through nutritional interventions such us use of probiotics, prebiotics or antibiotics, among others, seeking for a reversion of immunosuppression present in the tumor microenvironment (80). For example it has been found that oral administration of Bifidobacterium induces dendritic cell function and increases CD8+ T cell accumulation in the tumor microenvironment, improving response to anti-PDL1 antibody in mouse models of cancer (81). Despite some advances, caution is warranted as the ideal beneficial lung microbiota and how to change it as well as the potential risks of this modification are not yet known. For example, uncontrolled microbiota manipulation might induce detrimental inflammatory reactions that could worsen rather than improve cancer evolution and treatment efficacy.

Several studies have supported that the lung microbiota may play a key role in carcinogenesis and in response to chemotherapy and immunotherapy. Gut microbiome is the most studied so far. Lung microbiome is currently being studied to determine how and when does it affect lung cancer development and the best techniques to analyze it. New techniques based on microbiota-associated metabolic fingerprints can be useful to identify microbiota composition as recently developed for gut microbiota from lung cancer patients without requiring RNA analyses (82). Notably, it has been found that the metabolic profile in lungs from diseased patients, including lung cancer, significantly differs from that in healthy individuals (8385). Notably some studies have found that specific metabolites are enriched in respiratory airways of patients with different diseases and this profile correlates with the presence of specific bacterial species (8688).

These results indicate that characterization of lung metabolome could be used as a surrogate marker of changes in lung microbiota in order to provide an easier and more robust technology to use lung microbiota (or their products) as a biomarker for diagnosis and immunotherapy response in lung cancer. A recent study found that the presence of specific microbial metabolites in plasma correlated with anti-PD1 efficacy in lung cancer patients (89). Although similar studies have not been performed yet in airways, a recent study has shown that the profile of Volatile Organic Compounds in exhaled breath from lung cancer patients determined using an electronic nose predicts the efficacy of anti-PD1 treatment (90). Although future studies will be required to characterize the origin of these metabolites (host and/or microbial) and its correlation with changes in airway microbial composition and the response to anti-PD1 therapy, this approach would be very useful to implement the use of airway microbiota in clinical management of lung cancer and immunotherapy treatment.

Analysis of the upper respiratory tract microbiota as the source of the lung and gastric microbiota could be useful if the correlation between both (oral/upper respiratory tract and lung/gut) would be demonstrated. The sampling would be facilitated by less invasive techniques for the patient (nasal smears, saliva samples.), with the possibility of evaluating the changes over time.

The study of the microbiota is a field that has just made its way, offering opportunities and challenges for improving immunotherapy efficacy. Our group is conducting a pilot study in this direction in lung cancer patients receiving immunotherapy.

Outstanding Questions

1. Can we use antibiotics to safely modulate the microbiota? How can we modify resistant bacteria's composition and achieve the proper balance? What are the risks of microbiota manipulation?

2. What conditioning does a patient require prior to a fecal microbiota transplant? What is the proper composition for each individual?

3. Would it be useful to have an individualized microbiota bank?

4. Which barrier is primarily responsible for the loss of epithelial barrier in lung cancer? Are there specific markers that allow us to differentiate them?

Author Contributions

MO-D conceived the paper. MO-D, EG, DI, and JP draft the first version of the manuscript. MC, MG, RL, LM-L, AR-L, JRP, AS, IT, and AY commented and added extra information. All authors reviewed and approved the final version.

Funding

JP reported research funding from BMS and Gilead and speaker honoraria from Gilead and Pfizer. EG reported research funding from BMS and Gilead. JP and EG were funded by FEDER (Fondo Europeo de Desarrollo Regional, Gobierno de Aragón (Group B29_17R), Ministerio de Ciencia, Innovación e Universidades (MCNU), Agencia Estatal de Investigación (SAF2017-83120-C2-1-R), Fundacion Inocente Inocente, ASPANOA and Carrera de la Mujer de Monzón. JP was supported by ARAID Foundation. DI reported consultation honoraria from AbbVie, Amgen, AstraZeneca, BMS, Boehringer Ingelheim, Eli Lilly Oncology, F. Hoffmann-La Roche, Merck, MSD, Novartis, Pierre Fabre, Pfizer, and Takeda. DI reported speaker honoraria from Amgen, AstraZeneca, BMS, Boehringer Ingelheim, Eli Lilly Oncology, F. Hoffmann-La Roche, MSD, Novartis, Pierre Fabre, and Pfizer. DI reported research grant from AstraZeneca, BMS, F. Hoffmann-La Roche, MSD, and Pierre Fabre. The funder was not involved in the study design, collection, analysis, interpretation of data, the writing of this article or the decision to submit it for publication.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. (2018) 68:394–424. doi: 10.3322/caac.21492

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ, Wu Y-L, et al. Lung cancer: current therapies and new targeted treatments. Lancet. (2017) 389:299–311. doi: 10.1016/S0140-6736(16)30958-8

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Ordóñez-Mena JM, Schöttker B, Mons U, Jenab M, Freisling H, Bueno-de-Mesquita B, et al. Quantification of the smoking-associated cancer risk with rate advancement periods: meta-analysis of individual participant data from cohorts of the CHANCES consortium. BMC Med. (2016) 14:62. doi: 10.1186/s12916-016-0607-5

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Pabani A, Butts CA. Current landscape of immunotherapy for the treatment of metastatic non-small-cell lung cancer. Curr Oncol. (2018) 25(Suppl. 1):S94–102. doi: 10.3747/co.25.3750

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, et al. Overall survival with durvalumab after chemoradiotherapy in stage III NSCLC. N Engl J Med. (2018) 379:2342–50. doi: 10.1056/NEJMoa1809697

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus docetaxel in advanced nonsquamous non–small-cell lung cancer. N Engl J Med. (2015) 373:1627–39. doi: 10.1056/NEJMoa1507643

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Brahmer J, Reckamp KL, Baas P, Crinò L, Eberhardt WEE, Poddubskaya E, et al. Nivolumab versus docetaxel in advanced squamous-cell non–small-cell lung cancer. N Engl J Med. (2015) 373:123–35. doi: 10.1056/NEJMoa1504627

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Herbst RS, Baas P, Kim D-W, Felip E, Pérez-Gracia JL, Han J-Y, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. (2016) 387:1540–50. doi: 10.1016/S0140-6736(15)01281-7

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. (2017) 389:255–65. doi: 10.1016/S0140-6736(16)32517-X

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Brahmer JR, Rodríguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fülöp A, et al. Health-related quality-of-life results for pembrolizumab versus chemotherapy in advanced, PD-L1-positive NSCLC (KEYNOTE-024): a multicentre, international, randomised, open-label phase 3 trial. Lancet Oncology. (2017) 18:1600–9. doi: 10.1016/S1470-2045(17)30690-3

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Gandhi L, Rodríguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, et al. Pembrolizumab plus chemotherapy in metastatic non–small-cell lung cancer. N Engl J Med. (2018) 378:2078–92. doi: 10.1056/NEJMoa1801005

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Socinski MA, Jotte RM, Cappuzzo F, Orlandi F, Stroyakovskiy D, Nogami N, et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N Engl J Med. (2018) 378:2288–301. doi: 10.1056/NEJMoa1716948

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Hellmann MD, Ciuleanu T-E, Pluzanski A, Lee JS, Otterson GA, Audigier-Valette C, et al. Nivolumab plus Ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med. (2018) 378:2093–104. doi: 10.1056/NEJMoa1801946

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Horn L, Mansfield AS, Szczesna A, Havel L, Krzakowski M, Hochmair MJ, et al. First-line atezolizumab plus chemotherapy in extensive-stage small-cell lung cancer. N Engl J Med. (2018) 379:2220–9. doi: 10.1056/NEJMoa1809064

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Paz-Ares L, Dvorkin M, Chen Y, Reinmuth N, Hotta K, Trukhin D, et al. Durvalumab plus platinum–etoposide versus platinum–etoposide in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): a randomised, controlled, open-label, phase 3 trial. Lancet. (2019) 394:1929–39. doi: 10.1016/S0140-6736(19)32222-6

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Hirsch FR, McElhinny A, Stanforth D, Ranger-Moore J, Jansson M, Kulangara K, et al. PD-L1 immunohistochemistry assays for lung cancer: results from phase 1 of the blueprint PD-L1 IHC assay comparison project. J Thorac Oncol. (2017) 12:208–22. doi: 10.1016/j.jtho.2016.11.2228

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Goubet A-G, Daillère R, Routy B, Derosa L, Roberti PM, Zitvogel L. The impact of the intestinal microbiota in therapeutic responses against cancer. C R Biol. (2018) 341:284–9. doi: 10.1016/j.crvi.2018.03.004

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Samuelson DR, Welsh DA, Shellito JE. Regulation of lung immunity and host defense by the intestinal microbiota. Front Microbiol. (2015) 6:1085. doi: 10.3389/fmicb.2015.01085

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Sender R, Fuchs S, Milo R. Revised estimates for the number of human and bacteria cells in the body. PLOS Biol. (2016) 14:e1002533. doi: 10.1371/journal.pbio.1002533

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Costello EK, Lauber CL, Hamady M, Fierer N, Gordon JI, Knight R. Bacterial community variation in human body habitats across space and time. Science. (2009) 326:1694–7. doi: 10.1126/science.1177486

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Bhatt AP, Redinbo MR, Bultman SJ. The role of the microbiome in cancer development and therapy. CA Cancer J Clin. (2017) 67:326–44. doi: 10.3322/caac.21398

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Helmink BA, Khan MAW, Hermann A, Gopalakrishnan V, Wargo JA. The microbiome, cancer, and cancer therapy. Nat Med. (2019) 25:377–88. doi: 10.1038/s41591-019-0377-7

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Dickson RP, Erb-Downward JR, Huffnagle GB. The role of the bacterial microbiome in lung disease. Expert Rev Respir Med. (2013) 7:245–57. doi: 10.1586/ers.13.24

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Schwabe RF, Jobin C. The microbiome and cancer. Nat Rev Cancer. (2013) 13:800–12. doi: 10.1038/nrc3610

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Rojo D, Méndez-García C, Raczkowska BA, Bargiela R, Moya A, Ferrer M, et al. Exploring the human microbiome from multiple perspectives: factors altering its composition and function. FEMS Microbiol Rev. (2017) 41:453–78. doi: 10.1093/femsre/fuw046

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Moya A, Ferrer M. Functional redundancy-induced stability of gut microbiota subjected to disturbance. Trends Microbiol. (2016) 24:402–13. doi: 10.1016/j.tim.2016.02.002

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Ramírez-Labrada AG, Isla D, Artal A, Arias M, Rezusta A, Pardo J, et al. The influence of lung microbiota on lung carcinogenesis, immunity, and immunotherapy. Trends Cancer. (2020) 6:86–97. doi: 10.1016/j.trecan.2019.12.007

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Zitvogel L, Ma Y, Raoult D, Kroemer G, Gajewski TF. The microbiome in cancer immunotherapy: diagnostic tools and therapeutic strategies. Science. (2018) 359:1366–70. doi: 10.1126/science.aar6918

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Matysiak-Budnik T, Mégraud F. Helicobacter pylori infection and gastric cancer. Eu J Cancer. (2006) 42:708–16. doi: 10.1016/j.ejca.2006.01.020

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Toor D, Wsson MK, Kumar P, Karthikeyan G, Kaushik NK, Goel C, et al. Dysbiosis disrupts gut immune homeostasis and promotes gastric diseases. Int J Mol Sci. (2019) 20:2432. doi: 10.3390/ijms20102432

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci USA. (2007) 104:13780–5. doi: 10.1073/pnas.0706625104

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Zitvogel L, Daillère R, Roberti MP, Routy B, Kroemer G. Anticancer effects of the microbiome and its products. Nat Rev Microbiol. (2017) 15:465–78. doi: 10.1038/nrmicro.2017.44

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Belcheva A, Irrazabal T, Robertson SJ, Streutker C, Maughan H, Rubino S, et al. Gut microbial metabolism drives transformation of Msh2-deficient colon epithelial cells. Cell. (2014) 158:288–99. doi: 10.1016/j.cell.2014.04.051

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Maddi A, Sabharwal A, Violante T, Manuballa S, Genco R, Patnaik S, et al. The microbiome and lung cancer. J Thorac Dis. (2019) 11:280–91. doi: 10.21037/jtd.2018.12.88

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Segal LN, Clemente JC, Tsay J-CJ, Koralov SB, Keller BC, Wu BG, et al. Enrichment of the lung microbiome with oral taxa is associated with lung inflammation of a Th17 phenotype. Nat Microbiol. (2016) 1:16031. doi: 10.1038/nmicrobiol.2016.31

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Tsay J-CJ, Wu BG, Badri MH, Clemente JC, Shen N, Meyn P, et al. Airway microbiota is associated with upregulation of the PI3K pathway in lung cancer. Am J Respir Crit Care Med. (2018) 198:1188–98. doi: 10.1164/rccm.201710-2118OC

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Mendez R, Banerjee S, Bhattacharya SK, Banerjee S. Lung inflammation and disease: a perspective on microbial homeostasis and metabolism. IUBMB Life. (2019) 71:152–65. doi: 10.1002/iub.1969

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Sulaiman I, Wu BG, Li Y, Scott AS, Malecha P, Scaglione B, et al. Evaluation of the airway microbiome in nontuberculous mycobacteria disease. Eur Respir J. (2018) 52:1800810. doi: 10.1183/13993003.00810-2018

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Durack J, Huang YJ, Nariya S, Christian LS, Ansel KM, Beigelman A, et al. Bacterial biogeography of adult airways in atopic asthma. Microbiome. (2018) 6:104. doi: 10.1186/s40168-018-0487-3

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Tsay J-CJ, Segal LN. Could the sputum microbiota be a biomarker that predicts mortality after acute exacerbations of chronic obstructive pulmonary disease? Am J Respir Crit Care Med. (2019) 199:1175–6. doi: 10.1164/rccm.201811-2138ED

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Sears CL, Geis AL, Housseau F. Bacteroides fragilis subverts mucosal biology: from symbiont to colon carcinogenesis. J Clin Invest. (2014) 124:4166–72. doi: 10.1172/JCI72334

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Huang D, Su X, Yuan M, Zhang S, He J, Deng Q, et al. The characterization of lung microbiome in lung cancer patients with different clinicopathology. Am J Cancer Res. (2019) 9:2047–63.

PubMed Abstract | Google Scholar

43. Shirazi MSR, Al-Alo KZK, Al-Yasiri MH, Lateef ZM, Ghasemian A. Microbiome dysbiosis and predominant bacterial species as human cancer biomarkers. J Gastrointest Cancer. (2019) 51:725–8. doi: 10.1007/s12029-019-00311-z

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Maddi A, Scannapieco FA. Oral biofilms, oral and periodontal infections, and systemic disease. Am J Dent. (2013) 26:249–54.

PubMed Abstract | Google Scholar

45. Scher JU, Joshua V, Artacho A, Abdollahi-Roodsaz S, Öckinger J, Kullberg S, et al. The lung microbiota in early rheumatoid arthritis and autoimmunity. Microbiome. (2016) 4:60. doi: 10.1186/s40168-016-0206-x

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Huang C, Shi G. Smoking and microbiome in oral, airway, gut and some systemic diseases. J Transl Med. (2019) 17:225. doi: 10.1186/s12967-019-1971-7

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Jin C, Lagoudas GK, Zhao C, Bullman S, Bhutkar A, Hu B, et al. Commensal microbiota promote lung cancer development via γδ T cells. Cell. (2019) 21;176:998–1013.e16. doi: 10.1016/j.cell.2018.12.040

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Palucka AK, Coussens LM. The basis of oncoimmunology. Cell. (2016) 164:1233–47. doi: 10.1016/j.cell.2016.01.049

PubMed Abstract | CrossRef Full Text | Google Scholar

49. An S-Q, Warris A, Turner S. Microbiome characteristics of induced sputum compared to bronchial fluid and upper airway samples. Pediatr Pulmonol. (2018) 53:921–8. doi: 10.1002/ppul.24037

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Kovaleva OV, Romashin D, Zborovskaya IB, Davydov MM, Shogenov MS, Gratchev A. Human lung microbiome on the way to cancer. J Immunol Res. (2019) 2019:1394191. doi: 10.1155/2019/1394191

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Lee SH, Sung JY, Yong D, Chun J, Kim SY, Song JH, et al. Characterization of microbiome in bronchoalveolar lavage fluid of patients with lung cancer comparing with benign mass like lesions. Lung Cancer. (2016) 102:89–95. doi: 10.1016/j.lungcan.2016.10.016

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Morris A, Beck JM, Schloss PD, Campbell TB, Crothers K, Curtis JL, et al. Comparison of the respiratory microbiome in healthy nonsmokers and smokers. Am J Respir Crit Care Med. 2013 187:1067–75. doi: 10.1164/rccm.201210-1913OC

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Peters BA, Hayes RB, Goparaju C, Reid C, Pass HI, Ahn J. The microbiome in lung cancer tissue and recurrence-free survival. Cancer Epidemiol Biomarkers Prev. (2019) 28:731–40. doi: 10.1158/1055-9965.EPI-18-0966

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Elinav E, Garrett WS, Trinchieri G, Wargo J. The cancer microbiome. Nat Rev Cancer. (2019) 19:371–6. doi: 10.1038/s41568-019-0155-3

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Takahashi S, Tomita J, Nishioka K, Hisada T, Nishijima M. Development of a prokaryotic universal primer for simultaneous analysis of bacteria and archaea using next-generation sequencing. PLoS ONE. (2014) 9:e105592. doi: 10.1371/journal.pone.0105592

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Aguiar-Pulido V, Huang W, Suarez-Ulloa V, Cickovski T, Mathee K, Narasimhan G. Metagenomics, metatranscriptomics, and metabolomics approaches for microbiome analysis. Evol Bioinform Online. (2016) 12(Suppl. 1):5–16. doi: 10.4137/EBO.S36436

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Gori S, Inno A, Belluomini L, Bocus P, Bisoffi Z, Russo A, et al. Gut microbiota and cancer: how gut microbiota modulates activity, efficacy and toxicity of antitumoral therapy. Crit Rev Oncol Hematol. (2019) 143:139–47. doi: 10.1016/j.critrevonc.2019.09.003

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Horvat TZ, Adel NG, Dang T-O, Momtaz P, Postow MA, Callahan MK, et al. Immune-related adverse events, need for systemic immunosuppression, and effects on survival and time to treatment failure in patients with melanoma treated with ipilimumab at memorial sloan kettering cancer center. J Clin Oncol. (2015) 33:3193–8. doi: 10.1200/JCO.2015.60.8448

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Brahmer JR, Lacchetti C, Schneider BJ, Atkins MB, Brassil KJ, Caterino JM, et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American society of clinical oncology clinical practice guideline. J Clin Oncol. (2018) 36:1714–68. doi: 10.1200/JCO.2017.77.6385

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Chaput N, Lepage P, Coutzac C, Soularue E, Le Roux K, Monot C, et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol. (2017) 28:1368–79. doi: 10.1093/annonc/mdx108

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Teraoka S, Fujimoto D, Morimoto T, Kawachi H, Ito M, Sato Y, et al. Early immune-related adverse events and association with outcome in advanced non-small cell lung cancer patients treated with nivolumab: a prospective cohort study. J Thorac Oncol. (2017) 12:1798–805. doi: 10.1016/j.jtho.2017.08.022

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science. (2018) 359:91–7. doi: 10.1126/science.aan3706

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Derosa L, Hellmann MD, Spaziano M, Halpenny D, Fidelle M, Rizvi H, et al. Negative association of antibiotics on clinical activity of immune checkpoint inhibitors in patients with advanced renal cell and non-small-cell lung cancer. Ann Oncol. (2018) 29:1437–44. doi: 10.1093/annonc/mdy103

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Bingula R, Filaire M, Radosevic-Robin N, Bey M, Berthon J-Y, Bernalier-Donadille A, et al. Desired turbulence? gut-lung axis, immunity, and lung cancer. J Oncol. (2017) 2017:1–15. doi: 10.1155/2017/5035371

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Stiemsma LT, Turvey SE. Asthma and the microbiome: defining the critical window in early life. Allergy Asthma Clin Immunol. (2017) 13:3. doi: 10.1186/s13223-016-0173-6

PubMed Abstract | CrossRef Full Text | Google Scholar

66. McKenzie C, Tan J, Macia L, Mackay CR. The nutrition-gut microbiome-physiology axis and allergic diseases. Immunol Rev. (2017) 278:277–95. doi: 10.1111/imr.12556

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Zhang W-Q, Zhao S-K, Luo J-W, Dong X-P, Hao Y-T, Li H, et al. Alterations of fecal bacterial communities in patients with lung cancer. Am J Transl Res. (2018) 10:3171–85.

PubMed Abstract | Google Scholar

68. Mörkl S, Lackner S, Meinitzer A, Mangge H, Lehofer M, Halwachs B, et al. Gut microbiota, dietary intakes and intestinal permeability reflected by serum zonulin in women. Eur J Nutr. (2018) 57:2985–97. doi: 10.1007/s00394-018-1784-0

CrossRef Full Text | Google Scholar

69. Tripathi A, Lammers KM, Goldblum S, Shea-Donohue T, Netzel-Arnett S, Buzza MS, et al. Identification of human zonulin, a physiological modulator of tight junctions, as prehaptoglobin-2. Proc Natl Acad Sci USA. 2009 106:16799–804. doi: 10.1073/pnas.0906773106

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Wells JM, Brummer RJ, Derrien M, MacDonald TT, Troost F, Cani PD, et al. Homeostasis of the gut barrier and potential biomarkers. Am J Physiol Gastrointest Liver Physiol. (2017) 312:G171–93. doi: 10.1152/ajpgi.00048.2015

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Vancamelbeke M, Vermeire S. The intestinal barrier: a fundamental role in health and disease. Expert Rev Gastroenterol Hepatol. (2017) 11:821–34. doi: 10.1080/17474124.2017.1343143

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Miranda-Ribera A, Ennamorati M, Serena G, Cetinbas M, Lan J, Sadreyev RI, et al. Exploiting the zonulin mouse model to establish the role of primary impaired gut barrier function on microbiota composition and immune profiles. Front Immunol. (2019) 10:2233. doi: 10.3389/fimmu.2019.02233

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Odenwald MA, Turner JR. The intestinal epithelial barrier: a therapeutic target? Nat Rev Gastroenterol Hepatol. (2017) 14:9–21. doi: 10.1038/nrgastro.2016.169

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Lindheim L, Bashir M, Münzker J, Trummer C, Zachhuber V, Leber B, et al. Alterations in gut microbiome composition and barrier function are associated with reproductive and metabolic defects in women with polycystic ovary syndrome (PCOS): a pilot study. PLoS ONE. (2017) 12:e0168390. doi: 10.1371/journal.pone.0168390

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Ajamian M, Steer D, Rosella G, Gibson PR. Serum zonulin as a marker of intestinal mucosal barrier function: may not be what it seems. PLoS ONE. (2019) 14:e0210728. doi: 10.1371/journal.pone.0210728

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. (2018) 359:97–103. doi: 10.1126/science.aan4236

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Jin Y, Dong H, Xia L, Yang Y, Zhu Y, Shen Y, et al. The diversity of gut microbiome is associated with favorable responses to anti–programmed death 1 immunotherapy in Chinese patients with NSCLC. J Thorac Oncol. (2019) 14:1378–89. doi: 10.1016/j.jtho.2019.04.007

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Fessler JL, Gajewski TF. The microbiota: a new variable impacting cancer treatment outcomes. Clin Cancer Res. (2017) 23:3229–31. doi: 10.1158/1078-0432.CCR-17-0864

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Budden KF, Gellatly SL, Wood DLA, Cooper MA, Morrison M, Hugenholtz P, et al. Emerging pathogenic links between microbiota and the gut–lung axis. Nat Rev Microbiol. (2017) 15:55–63. doi: 10.1038/nrmicro.2016.142

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Le Noci V, Guglielmetti S, Arioli S, Camisaschi C, Bianchi F, Sommariva M, et al. Modulation of pulmonary microbiota by antibiotic or probiotic aerosol therapy: a strategy to promote immunosurveillance against lung metastases. Cell Rep. (2018) 24:3528–38. doi: 10.1016/j.celrep.2018.08.090

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, et al. Commensal bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. (2015) 350:1084–9. doi: 10.1126/science.aac4255

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Botticelli A, Vernocchi P, Marini F, Quagliariello A, Cerbelli B, Reddel S, et al. Gut metabolomics profiling of non-small cell lung cancer (NSCLC) patients under immunotherapy treatment. J Transl Med. (2020) 18:49. doi: 10.1186/s12967-020-02231-0

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Cameron SJS, Lewis KE, Beckmann M, Allison GG, Ghosal R, Lewis PD, et al. The metabolomic detection of lung cancer biomarkers in sputum. Lung Cancer. (2016) 94:88–95. doi: 10.1016/j.lungcan.2016.02.006

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Nambiar S, How SB, Gummer J, Trengove R, Moodley Y. Metabolomics in chronic lung diseases. Respirology. (2020) 25:139–48. doi: 10.1111/resp.13530

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Stringer KA, McKay RT, Karnovsky A, Quémerais B, Lacy P. Metabolomics and its application to acute lung diseases. Front Immunol. (2016) 7:44. doi: 10.3389/fimmu.2016.00044

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Cribbs SK, Uppal K, Li S, Jones DP, Huang L, Tipton L, et al. Correlation of the lung microbiota with metabolic profiles in bronchoalveolar lavage fluid in HIV infection. Microbiome. (2016) 4:3. doi: 10.1186/s40168-016-0147-4

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Gonçalves SM, Lagrou K, Duarte-Oliveira C, Maertens JA, Cunha C, Carvalho A. The microbiome-metabolome crosstalk in the pathogenesis of respiratory fungal diseases. Virulence. (2016) 8:673–84. doi: 10.1080/21505594.2016.1257458

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Sharma NS, Vestal G, Wille K, Patel KN, Cheng F, Tipparaju S, et al. Differences in airway microbiome and metabolome of single lung transplant recipients. Respir Res. (2020) 21:104. doi: 10.1186/s12931-020-01367-3

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Hatae R, Chamoto K, Kim YH, Sonomura K, Taneishi K, Kawaguchi S, et al. Combination of host immune metabolic biomarkers for the PD-1 blockade cancer immunotherapy. JCI Insight. (2020) 5:e133501. doi: 10.1172/jci.insight.133501

PubMed Abstract | CrossRef Full Text | Google Scholar

90. de Vries R, Muller M, van der Noort V, Theelen WSME, Schouten RD, Hummelink K, et al. Prediction of response to anti-PD-1 therapy in patients with non-small-cell lung cancer by electronic nose analysis of exhaled breath. Ann Oncol. (2019) 30:1660–6. doi: 10.1093/annonc/mdz279

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: microbiota, lung cancer, immunotherapy, lung microbiota, antibiotics

Citation: Ocáriz-Díez M, Cruellas M, Gascón M, Lastra R, Martínez-Lostao L, Ramírez-Labrada A, Paño JR, Sesma A, Torres I, Yubero A, Pardo J, Isla D and Gálvez EM (2020) Microbiota and Lung Cancer. Opportunities and Challenges for Improving Immunotherapy Efficacy. Front. Oncol. 10:568939. doi: 10.3389/fonc.2020.568939

Received: 17 June 2020; Accepted: 26 August 2020;
Published: 24 September 2020.

Edited by:

Kartik Sehgal, Beth Israel Deaconess Medical Center and Harvard Medical School, United States

Reviewed by:

James Tsay, New York University, United States
Marcello Tiseo, University Hospital of Parma, Italy

Copyright © 2020 Ocáriz-Díez, Cruellas, Gascón, Lastra, Martínez-Lostao, Ramírez-Labrada, Paño, Sesma, Torres, Yubero, Pardo, Isla and Gálvez. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Maitane Ocáriz-Díez, mocarizdiez@gmail.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.