Prospective correlation between the patient microbiome with response to and development of immune-mediated adverse effects to immunotherapy in lung cancer

Justin Chau, Meeta Yadav, Ben Liu, Muhammad Furqan, Qun Dai, Shailesh Shahi, Arnav Gupta, Keri Nace Mercer, Evan Eastman, Taher Abu Hejleh, Carlos Chan, George J Weiner, Catherine Cherwin, Sonny T M Lee, Cuncong Zhong, Ashutosh Mangalam, Jun Zhang, Justin Chau, Meeta Yadav, Ben Liu, Muhammad Furqan, Qun Dai, Shailesh Shahi, Arnav Gupta, Keri Nace Mercer, Evan Eastman, Taher Abu Hejleh, Carlos Chan, George J Weiner, Catherine Cherwin, Sonny T M Lee, Cuncong Zhong, Ashutosh Mangalam, Jun Zhang

Abstract

Background: Though the gut microbiome has been associated with efficacy of immunotherapy (ICI) in certain cancers, similar findings have not been identified for microbiomes from other body sites and their correlation to treatment response and immune related adverse events (irAEs) in lung cancer (LC) patients receiving ICIs.

Methods: We designed a prospective cohort study conducted from 2018 to 2020 at a single-center academic institution to assess for correlations between the microbiome in various body sites with treatment response and development of irAEs in LC patients treated with ICIs. Patients must have had measurable disease, ECOG 0-2, and good organ function to be included. Data was collected for analysis from January 2019 to October 2020. Patients with histopathologically confirmed, advanced/metastatic LC planned to undergo immunotherapy-based treatment were enrolled between September 2018 and June 2019. Nasal, buccal and gut microbiome samples were obtained prior to initiation of immunotherapy +/- chemotherapy, at development of adverse events (irAEs), and at improvement of irAEs to grade 1 or less.

Results: Thirty-seven patients were enrolled, and 34 patients were evaluable for this report. 32 healthy controls (HC) from the same geographic region were included to compare baseline gut microbiota. Compared to HC, LC gut microbiota exhibited significantly lower α-diversity. The gut microbiome of patients who did not suffer irAEs were found to have relative enrichment of Bifidobacterium (p = 0.001) and Desulfovibrio (p = 0.0002). Responders to combined chemoimmunotherapy exhibited increased Clostridiales (p = 0.018) but reduced Rikenellaceae (p = 0.016). In responders to chemoimmunotherapy we also observed enrichment of Finegoldia in nasal microbiome, and increased Megasphaera but reduced Actinobacillus in buccal samples. Longitudinal samples exhibited a trend of α-diversity and certain microbial changes during the development and resolution of irAEs.

Conclusions: This pilot study identifies significant differences in the gut microbiome between HC and LC patients, and their correlation to treatment response and irAEs in LC. In addition, it suggests potential predictive utility in nasal and buccal microbiomes, warranting further validation with a larger cohort and mechanistic dissection using preclinical models.

Trial registration: ClinicalTrials.gov, NCT03688347 . Retrospectively registered 09/28/2018.

Keywords: Adverse effects; Immune checkpoint; Immunotherapy; Lung cancer; Microbiome; Response; Toxicity.

Conflict of interest statement

The authors of this manuscript declare no conflicts of interest.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Description of patient cohorts and study schema. (a) Study schema showing the collection of microbiome samples from three separate body sites. Samples undergo 16S rRNA amplicon sequencing followed by taxonomic profiling. The resulting data is correlated to clinical outcomes such as response to ICI therapy or development of AEs. (b) An abbreviated demographics chart summarizing notable disease and patient characteristics of LC contributors. For granular individual characteristics, please refer to Supplemental Table 1. (c) Breakdown of patients belonging to each study cohort. ICI = immune checkpoint inhibitor. Checkpoint inhibitor status (number of patients enrolled – number of fecal samples that were unable to be analyzed or not submitted – number of nasal/buccal samples that were unable to be analyzed or not submitted
Fig. 2
Fig. 2
Flowchart of patients enrolled in the study. Breakdown of number of patients enrolled, were deemed ineligible for the study, as well as number of samples provided for each stage of analysis
Fig. 3
Fig. 3
Baseline microbiome composition. Bar and heatmap plots comparing baseline gut, nasal and buccal microbiomes in LC compared to HC. (a)Left: Bar graph showing relative ratios of phyla constitution in LC and HC samples. Right: Box plot showing a statistically significant decrease in α-diversity when comparing LC to HC patients (p = 9.36 × 10− 04). (b)Left: 2-dimensional PLS-DA graph identifying notable differences in genus expression when comparing LC vs HC samples. Right: Heatmap showing genus level expression in LC patients (upper half) compared with HC (lower half). There is a notable difference at the genus level. (c) Principal coordinate analysis (PCoA) comparing the beta-diversity of buccal, nasal and gut microbiome in LC patients
Fig. 4
Fig. 4
Response to immunotherapy. Microbiome changes notable in responders to ICI. Normalized data is presented in log-adjusted relative abundances. Left panels show PLS-DA graphs from nasal, buccal and gut sites all showing a microbiome separation when comparing ICI responders vs nonresponders. (a) Taxonomic profiling of nasal samples identified notable enrichment in Finegoldia, of phylum Firmicutes, in responders to ICI (p = 0.0005). (b) Buccal analysis of ICI responders show enrichment in Megasphaera of phylum Firmicutes (p = 8.6 × 10− 03) and decrease in Actinobacillus of phylum Proteobacteria (p = 9.7 × 10− 03). (c) In the fecal samples of ICI responders, Clostridiales was enriched (phylum Firmicutes, p = 0.017875) and Rikenellaceae decreased (phylum Bacteroidetes, p = 0.016013)
Fig. 5
Fig. 5
Toxicity analysis. (a) PLS-DA analysis showing significant microbiome differences between LC patients who experienced toxicities and those who did not using different grouping methods of irAE severities, e.g. grade 0 vs. grade 1 + 2 + 3 + 4; grade 0 + 1 vs. grade 2 + 3 + 4; and grade 0 vs. 1 + 2 vs. 3 + 4. (b) Normalized abundances of Bifidobacterium (phylum Actinobacteria) and Desulfovibrio (phylum Proteobacteria) showed enrichment in both bacteria in patients who developed less irAEs. All differences were statistically significant irrespective of categorization of AE severity
Fig. 6
Fig. 6
Longitudinal changes in microbiome with development and resolution of toxicities. (a) Analysis identified five patients who had submitted multiple samples during development and resolution of irAE. JZLC-24 and JZLC-6 did not have stool samples available for analysis but did submit all three sets of nasal and buccal samples. On the x-axis, sample collections are listed: V1, prior to initiation of immunotherapy; V2; at onset of toxicity, and V3, at resolution of irAE. The y-axis denotes the logarithmic (base 10) relative change in α -diversity compared to the previous visit, trended by the line plots, overlaid. Across nearly all sets of microbiome samples, a drop in microbiome α-diversity is observed at onset of irAE. At resolution of irAE to grade 1 severity or better, a third set of samples exhibit a trend toward either slowed rate of decrease in α-diversity or a reversal altogether toward baseline. (b) A consistent trend in increase of Staphylococcus at onset of toxicity and decrease with resolution of toxicity was also observed in the nasal samples (left), with a similar trend in buccal samples (right). (c)Megasphaera, a bacterium belonging to Firmicutes, was previously identified as being enriched in responders to immunotherapy. Here, it is also shown to decrease in buccal samples of patients who developed irAEs, then increasing in abundance with resolution of toxicity

References

    1. Swami U, Zakharia Y, Zhang J. Understanding microbiome Effect on immune checkpoint inhibition in lung Cancer: placing the puzzle pieces together. J Immunother. 2018;41(8):359–360. doi: 10.1097/CJI.0000000000000232.
    1. Strouse C, Mangalam A, Zhang J. Bugs in the system: bringing the human microbiome to bear in cancer immunotherapy. Gut Microbes. 2019;10(2):109–112. doi: 10.1080/19490976.2018.1511665.
    1. Temraz S, Nassar F, Nasr R, Charafeddine M, Mukherji D, Shamseddine A. Gut Microbiome: A Promising Biomarker for Immunotherapy in Colorectal Cancer. Int J Mol Sci. 2019;20(17). 10.3390/ijms20174155.
    1. Liong MT. Roles of probiotics and prebiotics in colon cancer prevention: postulated mechanisms and in-vivo evidence. Int J Mol Sci. 2008;9(5):854–863. doi: 10.3390/ijms9050854.
    1. Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell. 2014;157(1):121–141. doi: 10.1016/j.cell.2014.03.011.
    1. Zhang H, Garcia Rodriguez LA, Hernandez-Diaz S. Antibiotic use and the risk of lung cancer. Cancer Epidemiol Biomark Prev. 2008;17(6):1308–1315. doi: 10.1158/1055-9965.EPI-07-2817.
    1. Sacksteder MR. Occurrence of spontaneous tumors in the germfree F344 rat. J Natl Cancer Inst. 1976;57(6):1371–1373. doi: 10.1093/jnci/57.6.1371.
    1. Laqueur G, Matsumoto H, Yamamoto R. Comparison of the carcinogenicity of Methylazoxymethanol-B-D-glucosiduronic acid in conventional and germfree Sprague-Dawley rats. J Natl Cancer Inst. 1981;67(5):1053–1055.
    1. Maddi A, Sabharwal A, Violante T, et al. The microbiome and lung cancer. J Thorac Dis. 2019;11(1):280–291. doi: 10.21037/jtd.2018.12.88.
    1. Zhang D, Li S, Wang N, Tan HY, Zhang Z, Feng Y. The cross-talk between gut microbiota and lungs in common lung diseases. Front Microbiol. 2020;11:301. doi: 10.3389/fmicb.2020.00301.
    1. Keely S, Talley NJ, Hansbro PM. Pulmonary-intestinal cross-talk in mucosal inflammatory disease. Mucosal Immunol. 2012;5(1):7–18. doi: 10.1038/mi.2011.55.
    1. Abrahamsson TR, Jakobsson HE, Andersson AF, Bjorksten B, Engstrand L, Jenmalm MC. Low gut microbiota diversity in early infancy precedes asthma at school age. Clin Exp Allergy. 2014;44(6):842–850. doi: 10.1111/cea.12253.
    1. Belkaid Y, Harrison OJ. Homeostatic Immunity and the Microbiota. Immunity. 2017;46(4):562–576. doi: 10.1016/j.immuni.2017.04.008.
    1. Dang AT, Marsland BJ. Microbes, metabolites, and the gut-lung axis. Mucosal Immunol. 2019;12(4):843–850. doi: 10.1038/s41385-019-0160-6.
    1. Jin C, Lagoudas GK, Zhao C, et al. Commensal Microbiota Promote Lung Cancer Development via gammadelta T Cells. Cell. 2019;176(5):998–1013 e16. doi: 10.1016/j.cell.2018.12.040.
    1. Paulos CM, Wrzesinski C, Kaiser A, et al. Microbial translocation augments the function of adoptively transferred self/tumor-specific CD8+ T cells via TLR4 signaling. J Clin Invest. 2007;117(8):2197–2204. doi: 10.1172/JCI32205.
    1. Pitt JM, Vetizou M, Waldschmitt N, et al. Fine-Tuning Cancer Immunotherapy: Optimizing the Gut Microbiome. Cancer Res. 2016;76(16):4602–4607. doi: 10.1158/0008-5472.CAN-16-0448.
    1. Vetizou M, Pitt JM, Daillere R, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350(6264):1079–1084. doi: 10.1126/science.aad1329.
    1. Sivan A, Corrales L, Hubert N, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350(6264):1084–1089. doi: 10.1126/science.aac4255.
    1. Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–97. doi: 10.1126/science.aan3706.
    1. Chaput N, Lepage P, Coutzac C, et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol. 2017;28(6):1368–1379. doi: 10.1093/annonc/mdx108.
    1. Gopalakrishnan V, Spencer CN, Nezi L, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97–103. doi: 10.1126/science.aan4236.
    1. Shahi SK, Zarei K, Guseva NV, Mangalam AK. Microbiota Analysis Using Two-step PCR and Next-generation 16S rRNA Gene Sequencing. J Vis Exp. 2019;(152). 10.3791/59980.
    1. Yilmaz P, Parfrey LW, Yarza P, Gerken J, Pruesse E, Quast C, Schweer T, Peplies J, Ludwig W, Glöckner FO. The SILVA and “all-species living tree project (LTP)” taxonomic frameworks. Nucleic Acids Res. 2014;42(D1):D643–D648. doi: 10.1093/nar/gkt1209.
    1. Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJ, Holmes SP. DADA2: high-resolution sample inference from Illumina amplicon data. Nat Methods. 2016;13(7):581–583. doi: 10.1038/nmeth.3869.
    1. Chong J, Liu P, Zhou G, Xia J. Using MicrobiomeAnalyst for comprehensive statistical, functional, and meta-analysis of microbiome data. Nat Protocol. 2020;15(3):799–821. doi: 10.1038/s41596-019-0264-1.
    1. Arndt D, Xia J, Liu Y, Zhou Y, Guo AC, Cruz JA, Sinelnikov I, Budwill K, Nesbo CL, Wishart DS. METAGENassist: a comprehensive web server for comparative metagenomics. Nucleic Acids Res. 2012;40(W1):W88–W95. doi: 10.1093/nar/gks497.
    1. Antonia SJ, Villegas A, Daniel D, et al. Durvalumab after Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. N Engl J Med. 2017. 10.1056/NEJMoa1709937.
    1. Paz-Ares L, Dvorkin M, Chen Y, 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. 10.1016/S0140-6736(19)32222-6.
    1. Biragyn A, Ferrucci L. Gut dysbiosis: a potential link between increased cancer risk in ageing and inflammaging. Lancet Oncol. 2018;19(6):e295–e304. doi: 10.1016/s1470-2045(18)30095-0.
    1. Chen J, Chia N, Kalari KR, et al. Multiple sclerosis patients have a distinct gut microbiota compared to healthy controls. Sci Rep. 2016;6:28484. doi: 10.1038/srep28484.
    1. Chau JJ, Yadav M, Furqan M, et al. World Conference on Lung Cancer. Barcelona: International Association for the Study of Lung Cancer; 2019. Analysis of patient microbiome and its correlation to immunotherapy response and toxicity in lung cancer. Abstract.
    1. Zhang J. Analysis of patient microbiome and its correlation to immunotherapy response and toxicity in lung Cancer. 2019.
    1. Zhang WQ, Zhao SK, Luo JW, Dong XP, Hao YT, Li H, Shan L, Zhou Y, Shi HB, Zhang ZY, Peng CL, Zhao XG. Alterations of fecal bacterial communities in patients with lung cancer. Am J Transl Res. 2018;10(10):3171–3185.
    1. Wu X, Zhang T, Chen X, Ji G, Zhang F. Microbiota transplantation: targeting cancer treatment. Cancer Lett. 2019;452:144–151. doi: 10.1016/j.canlet.2019.03.010.
    1. Gong J, Chehrazi-Raffle A, Placencio-Hickok V, Guan M, Hendifar A, Salgia R. The gut microbiome and response to immune checkpoint inhibitors: preclinical and clinical strategies. Clin Transl Med. 2019;8(1):9. doi: 10.1186/s40169-019-0225-x.
    1. Jin Y, Dong H, Xia L, Yang Y, Zhu Y, Shen Y, Zheng H, Yao C, Wang Y, Lu S. 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(8):1378–1389. doi: 10.1016/j.jtho.2019.04.007.
    1. Huang C, Li M, Liu B, et al. Relating Gut Microbiome and Its Modulating Factors to Immunotherapy in Solid Tumors: A Systematic Review. Systematic Review. Front Oncol. 2021;11(760). 10.3389/fonc.2021.642110.
    1. Luu M, Weigand K, Wedi F, et al. Regulation of the effector function of CD8(+) T cells by gut microbiota-derived metabolite butyrate. Sci Rep. 2018;8(1):14430. doi: 10.1038/s41598-018-32860-x.
    1. Trompette A, Gollwitzer ES, Pattaroni C, et al. Dietary Fiber Confers Protection against Flu by Shaping Ly6c(−) Patrolling Monocyte Hematopoiesis and CD8(+) T Cell Metabolism. Immunity. 2018;48(5):992–1005 e8. doi: 10.1016/j.immuni.2018.04.022.
    1. Park J, Kim M, Kang SG, Jannasch AH, Cooper B, Patterson J, Kim CH. Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR-S6K pathway. Mucosal Immunol. 2015;8(1):80–93. doi: 10.1038/mi.2014.44.
    1. Byndloss MX, Olsan EE, Rivera-Chavez F, et al. Microbiota-activated PPAR-gamma signaling inhibits dysbiotic Enterobacteriaceae expansion. Science. 2017;357(6351):570–575. doi: 10.1126/science.aam9949.
    1. Litvak Y, Byndloss MX, Baumler AJ. Colonocyte metabolism shapes the gut microbiota. Science. 2018;362(6418). 10.1126/science.aat9076.
    1. Atarashi K, Tanoue T, Ando M, et al. Th17 Cell Induction by Adhesion of Microbes to Intestinal Epithelial Cells. Cell. 2015;163(2):367–380. doi: 10.1016/j.cell.2015.08.058.
    1. Suarez G, Romero-Gallo J, Piazuelo MB, et al. Nod1 Imprints Inflammatory and Carcinogenic Responses toward the Gastric Pathogen Helicobacter pylori. Cancer Res. 2019;79(7):1600–1611. doi: 10.1158/0008-5472.CAN-18-2651.
    1. Tran LS, Tran D, De Paoli A, et al. NOD1 is required for helicobacter pylori induction of IL-33 responses in gastric epithelial cells. Cell Microbiol. 2018;20(5):e12826. doi: 10.1111/cmi.12826.
    1. Lehours P, Ferrero RL. Review: Helicobacter: Inflammation, immunology, and vaccines. Helicobacter. 2019;24(Suppl 1):e12644. doi: 10.1111/hel.12644.
    1. Frankel AE, Coughlin LA, Kim J, Froehlich TW, Xie Y, Frenkel EP, Koh AY. Metagenomic shotgun sequencing and unbiased Metabolomic profiling identify specific human gut microbiota and metabolites associated with immune checkpoint therapy efficacy in melanoma patients. Neoplasia. 2017;19(10):848–855. doi: 10.1016/j.neo.2017.08.004.
    1. Huang C, Li M, Liu B, et al. Relating Gut Microbiome and Its Modulating Factors to Immunotherapy in Solid Tumors: A Systematic Review. Research Square. 2020. 10.21203/-73124/v1.
    1. Wang F, Yin Q, Chen L, Davis MM. Bifidobacterium can mitigate intestinal immunopathology in the context of CTLA-4 blockade. Proc Natl Acad Sci U S A. 2018;115(1):157–161. doi: 10.1073/pnas.1712901115.
    1. McGeachy MJ, Cua DJ, Gaffen SL. The IL-17 Family of Cytokines in Health and Disease. Immunity. 2019;50(4):892–906. doi: 10.1016/j.immuni.2019.03.021.
    1. Schirmer M, Smeekens SP, Vlamakis H, et al. Linking the Human Gut Microbiome to Inflammatory Cytokine Production Capacity. Cell. 2016;167(4):1125–1136 e8. doi: 10.1016/j.cell.2016.10.020.
    1. Warren YA, Citron DM, Merriam CV, Goldstein EJ. Biochemical differentiation and comparison of Desulfovibrio species and other phenotypically similar genera. J Clin Microbiol. 2005;43(8):4041–4045. doi: 10.1128/JCM.43.8.4041-4045.2005.
    1. Rowan F, Docherty NG, Murphy M, Murphy B, Calvin Coffey J, O'Connell PR. Desulfovibrio bacterial species are increased in ulcerative colitis. Dis Colon rectum. 2010;53(11):1530–1536. doi: 10.1007/DCR.0b013e3181f1e620.
    1. Dzierzewicz Z, Cwalina B, Kuśmierz D, Kwapisz I. Differences between Desulfovibrio desulfuricans intestinal strains with respect to their ability of sulphasalazine biotransformation. Acta Pol Pharm. 2000;57:124–126.
    1. Wilson ID, Nicholson JK. Gut microbiome interactions with drug metabolism, efficacy, and toxicity. Transl Res. 2017;179:204–222. doi: 10.1016/j.trsl.2016.08.002.
    1. Greathouse KL, White JR, Vargas AJ, et al. Interaction between the microbiome and TP53 in human lung cancer. Genome Biol. 2018;19(1):123. doi: 10.1186/s13059-018-1501-6.
    1. Hekmatshoar Y, Rahbar Saadat Y, Hosseiniyan Khatibi SM, et al. The impact of tumor and gut microbiotas on cancer therapy: Beneficial or detrimental? Life Sci. 2019;233:116680. doi: 10.1016/j.lfs.2019.116680.
    1. Otto M. Staphylococci in the human microbiome: the role of host and interbacterial interactions. Curr Opin Microbiol. 2020;53:71–77. doi: 10.1016/j.mib.2020.03.003.
    1. Ceccarelli F, Perricone C, Olivieri G, et al. Staphylococcus aureus Nasal Carriage and Autoimmune Diseases: From Pathogenic Mechanisms to Disease Susceptibility and Phenotype. Int J Mol Sci. 2019;20(22). 10.3390/ijms20225624.
    1. Helmink BA, Khan MAW, Hermann A, Gopalakrishnan V, Wargo JA. The microbiome, cancer, and cancer therapy. Nat Med. 2019;25(3):377–388. doi: 10.1038/s41591-019-0377-7.
    1. Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, Luke JJ, Gajewski TF. The commensal microbiome is associated with anti–PD-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104–108. doi: 10.1126/science.aao3290.
    1. Stangroom J. Effect Size Calculator for T-Test. Jeremy Stangroom. May 15, 2021. 2021.
    1. Bullman S. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science. 2017;358(6369):1443–1448. doi: 10.1126/science.aal5240.
    1. Yu T, Guo F, Yu Y, et al. Fusobacterium nucleatum Promotes Chemoresistance to Colorectal Cancer by Modulating Autophagy. Cell. 2017;170(3):548–563 e16. doi: 10.1016/j.cell.2017.07.008.
    1. Zorron Cheng Tao Pu L, Yamamoto K, Honda T, Nakamura M, Yamamura T, Hattori S, Burt AD, Singh R, Hirooka Y, Fujishiro M. Microbiota profile is different for early and invasive colorectal cancer and is consistent throughout the colon. J Gastroenterol Hepatol. 2020;35(3):433–437. doi: 10.1111/jgh.14868.

Source: PubMed

3
订阅