Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

A non-antibiotic-disrupted gut microbiome is associated with clinical responses to CD19-CAR-T cell cancer immunotherapy

Abstract

Increasing evidence suggests that the gut microbiome may modulate the efficacy of cancer immunotherapy. In a B cell lymphoma patient cohort from five centers in Germany and the United States (Germany, n = 66; United States, n = 106; total, n = 172), we demonstrate that wide-spectrum antibiotics treatment (‘high-risk antibiotics’) prior to CD19-targeted chimeric antigen receptor (CAR)-T cell therapy is associated with adverse outcomes, but this effect is likely to be confounded by an increased pretreatment tumor burden and systemic inflammation in patients pretreated with high-risk antibiotics. To resolve this confounding effect and gain insights into antibiotics-masked microbiome signals impacting CAR-T efficacy, we focused on the high-risk antibiotics non-exposed patient population. Indeed, in these patients, significant correlations were noted between pre-CAR-T infusion Bifidobacterium longum and microbiome-encoded peptidoglycan biosynthesis, and CAR-T treatment-associated 6-month survival or lymphoma progression. Furthermore, predictive pre-CAR-T treatment microbiome-based machine learning algorithms trained on the high-risk antibiotics non-exposed German cohort and validated by the respective US cohort robustly segregated long-term responders from non-responders. Bacteroides, Ruminococcus, Eubacterium and Akkermansia were most important in determining CAR-T responsiveness, with Akkermansia also being associated with pre-infusion peripheral T cell levels in these patients. Collectively, we identify conserved microbiome features across clinical and geographical variations, which may enable cross-cohort microbiome-based predictions of outcomes in CAR-T cell immunotherapy.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Association of antibiotic drug exposure prior to CD19-directed CAR-T cell therapy with survival outcomes, tumor burden and inflammation.
Fig. 2: Gut microbiome characteristics and association with clinical variables in CAR-T cell-treated patients.
Fig. 3: Prediction of response to CAR-T cell therapy by specific gut commensals.
Fig. 4: Association of gut microbes with patient T cells before CAR-T cell infusion.

Similar content being viewed by others

Data availability

All data are available in the main text or the extended materials. The sequencing data generated during this study are available at European Nucleotide Archive (ENA) (project accession no. PRJEB54704) including basic phenotypes.

Code availability

Code used in the present study is available at https://github.com/Elinav-Lab-DKFZ/CART-Microbiome and upon request.

References

  1. Gopalakrishnan, V. et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 359, 97–103 (2018).

    CAS  PubMed  Google Scholar 

  2. Routy, B. et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 359, 91–97 (2018).

    CAS  PubMed  Google Scholar 

  3. Matson, V. et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 359, 104–108 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Peled, J. U. et al. Intestinal microbiota and relapse after hematopoietic-cell transplantation. J. Clin. Oncol. 35, 1650–1659 (2017).

    PubMed  PubMed Central  Google Scholar 

  5. Stein-Thoeringer, C. K. et al. Lactose drives Enterococcus expansion to promote graft-versus-host disease. Science 366, 1143–1149 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Mager, L. F. et al. Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy. Science 369, 1481–1489 (2020).

    CAS  PubMed  Google Scholar 

  7. Davar, D. et al. Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science 371, 595–602 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Baruch, E. N. et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science 371, 602–609 (2021).

    CAS  PubMed  Google Scholar 

  9. Pinato, D. J. et al. Association of prior antibiotic treatment with survival and response to immune checkpoint inhibitor therapy in patients with cancer. JAMA Oncol. 5, 1774–1778 (2019).

    PubMed  PubMed Central  Google Scholar 

  10. Shono, Y. et al. Increased GVHD-related mortality with broad-spectrum antibiotic use after allogeneic hematopoietic stem cell transplantation in human patients and mice. Sci. Transl. Med. 8, 339ra371 (2016).

    Google Scholar 

  11. Guedan, S., Ruella, M. & June, C. H. Emerging cellular therapies for cancer. Annu. Rev. Immunol. 37, 145–171 (2019).

    CAS  PubMed  Google Scholar 

  12. Amini, L. et al. Preparing for CAR T cell therapy: patient selection, bridging therapies and lymphodepletion. Nat. Rev. Clin. Oncol. 19, 342–355 (2022).

    PubMed  Google Scholar 

  13. Park, J. H. et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N. Engl. J. Med. 378, 449–459 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Locke, F. L. et al. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1-2 trial. Lancet Oncol. 20, 31–42 (2019).

    CAS  PubMed  Google Scholar 

  15. Nastoupil, L. J. et al. Standard-of-care axicabtagene ciloleucel for relapsed or refractory large B-cell lymphoma: results from the US Lymphoma CAR T consortium. J. Clin. Oncol. 38, 3119–3128 (2020).

    PubMed  PubMed Central  Google Scholar 

  16. Schuster, S. J. et al. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N. Engl. J. Med. 380, 45–56 (2019).

    CAS  PubMed  Google Scholar 

  17. Jain, M. D. et al. Tumor interferon signaling and suppressive myeloid cells are associated with CAR T-cell failure in large B-cell lymphoma. Blood 137, 2621–2633 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Deng, Q. et al. Characteristics of anti-CD19 CAR T cell infusion products associated with efficacy and toxicity in patients with large B cell lymphomas. Nat. Med. 26, 1878–1887 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Plaks, V. et al. CD19 target evasion as a mechanism of relapse in large B-cell lymphoma treated with axicabtagene ciloleucel. Blood 138, 1081–1085 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Jain, M. D. et al. Whole-genome sequencing reveals complex genomic features underlying anti-CD19 CAR T-cell treatment failures in lymphoma. Blood 140, 491–503 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Dean, E. A. et al. High metabolic tumor volume is associated with decreased efficacy of axicabtagene ciloleucel in large B-cell lymphoma. Blood Adv. 4, 3268–3276 (2020). `.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Karschnia, P. et al. Clinical presentation, management, and biomarkers of neurotoxicity after adoptive immunotherapy with CAR T cells. Blood 133, 2212–2221 (2019).

    CAS  PubMed  Google Scholar 

  23. Jacobson, C. A. et al. Axicabtagene ciloleucel in the non-trial setting: outcomes and correlates of response, resistance, and toxicity. J. Clin. Oncol. 38, 3095–3106 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Smith, M. et al. Gut microbiome correlates of response and toxicity following anti-CD19 CAR T cell therapy. Nat. Med. 28, 713–723 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Cullin, N., Azevedo Antunes, C., Straussman, R., Stein-Thoeringer, C. K. & Elinav, E. Microbiome and cancer. Cancer Cell 39, 1317–1341 (2021).

    CAS  PubMed  Google Scholar 

  26. Galloway-Pena, J. R. et al. Gut microbiome signatures are predictive of infectious risk following induction therapy for acute myeloid leukemia. Clin. Infect. Dis. 71, 63–71 (2020).

    CAS  PubMed  Google Scholar 

  27. Santomasso, B., Bachier, C., Westin, J., Rezvani, K. & Shpall, E. J. The other side of CAR T-cell therapy: cytokine release syndrome, neurologic toxicity, and financial burden. Am. Soc. Clin. Oncol. Educ. Book 39, 433–444 (2019).

    PubMed  Google Scholar 

  28. Derosa, L. 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. 29, 1437–1444 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Bachy, E. et al. A real-world comparison of tisagenlecleucel and axicabtagene ciloleucel CAR T cells in relapsed or refractory diffuse large B cell lymphoma. Nat. Med. 28, 2145–2154 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Finney, O. C. et al. CD19 CAR T cell product and disease attributes predict leukemia remission durability. J. Clin. Invest. 129, 2123–2132 (2019).

    PubMed  PubMed Central  Google Scholar 

  31. Das, R. K., Vernau, L., Grupp, S. A. & Barrett, D. M. Naive T-cell deficits at diagnosis and after chemotherapy impair cell therapy potential in pediatric cancers. Cancer Discov. 9, 492–499 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Gacesa, R. et al. Environmental factors shaping the gut microbiome in a Dutch population. Nature 604, 732–739 (2022).

    CAS  PubMed  Google Scholar 

  33. Schluter, J. et al. The gut microbiota is associated with immune cell dynamics in humans. Nature 588, 303–307 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Wastyk, H. C. et al. Gut-microbiota-targeted diets modulate human immune status. Cell 184, 4137–4153 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Castello, A., Rossi, S., Toschi, L. & Lopci, E. Impact of antibiotic therapy and metabolic parameters in non-small cell lung cancer patients receiving checkpoint inhibitor. J. Clin. Med. 10, 1251 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Lee, K. A. et al. Cross-cohort gut microbiome associations with immune checkpoint inhibitor response in advanced melanoma. Nat. Med. 28, 535–544 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Cammarota, G. et al. Gut microbiome, big data and machine learning to promote precision medicine for cancer. Nat. Rev. Gastroenterol. Hepatol. 17, 635–648 (2020).

    PubMed  Google Scholar 

  38. McCulloch, J. A. et al. Intestinal microbiota signatures of clinical response and immune-related adverse events in melanoma patients treated with anti-PD-1. Nat. Med. 28, 545–556 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Dizman, N. et al. Nivolumab plus ipilimumab with or without live bacterial supplementation in metastatic renal cell carcinoma: a randomized phase 1 trial. Nat. Med. 28, 704–712 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Derrien, M., Vaughan, E. E., Plugge, C. M. & de Vos, W. M. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int. J. Syst. Evol. Microbiol. 54, 1469–1476 (2004).

    CAS  PubMed  Google Scholar 

  41. Luu, M. et al. Microbial short-chain fatty acids modulate CD8(+) T cell responses and improve adoptive immunotherapy for cancer. Nat. Commun. 12, 4077 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Bae, M. et al. Akkermansia muciniphila phospholipid induces homeostatic immune responses. Nature 608, 168–173 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Gamage, H. et al. Third generation cephalosporins and piperacillin/tazobactam have distinct impacts on the microbiota of critically ill patients. Sci. Rep. 11, 7252 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Lee, D. W. et al. ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol. Blood Marrow Transplant. 25, 625–638 (2019).

    CAS  PubMed  Google Scholar 

  45. Hogue, S. R., Gomez, M. F., da Silva, W. V. & Pierce, C. M. A customized at-home stool collection protocol for use in microbiome studies conducted in cancer patient populations. Microb. Ecol. 78, 1030–1034 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Beghini, F. et al. Integrating taxonomic, functional, and strain-level profiling of diverse microbial communities with bioBakery 3. Elife 10, e65088 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Hastie, T., Tibshirani, R. & Friedman, J. The Elements of Statistical Learning: Data Mining, Inference, and Prediction 2nd edn (Springer, 2009).

  48. McElreath, R. A Bayesian Course with Examples in R and STAN 2nd edn (Chapman & Hall, 2020).

  49. Bingham, E. et al. Pyro: deep universal probabilistic programming. Journal of Machine Learning Research 20, 973–978 (2019).

    Google Scholar 

  50. Wingate, D. & Weber, T. Automated variational inference in probabilistic programming. Preprint at https://doi.org/10.48550/arxiv.1301.1299 (2013).

Download references

Acknowledgements

We thank the members of the DKFZ Microbiome & Cancer Division and the Elinav laboratory, Weizmann Institute of Science for insightful discussions; and the participants, investigators, collaborators and study site personnel engaging in the trials. We also thank the REG allo-SCT/CAR team, especially H. Bremm, T. Schifferstein and Y. Schumann for their help in collecting and cryopreserving stool samples, and S. Gleich for data management. We are grateful for the partnership with the Mark Foundation (Endeavor Award 2021 to C.K.S.-T., E.E., R.R.J., M.D.J., M.L.D.), which funded key aspects of this study. C.K.S.-T. is supported by the German José Carreras Leukemia Foundation (01 R/2020), the German Research Foundation (DFG; STE 2964/5-1), the Baden-Württemberg Stiftung (MiCART19), the DFG Excellence Cluster EXC-2124 (Controlling Microbes to Fight Infections, CMFI) and the NCT Heidelberg Funds against Cancer. V.B. is supported by the Else-Kröner Fresenius-Stiftung and the German Cancer Consortium (DKTK) and the Bavarian Cancer for Cancer Research (BZKF). M.-L.S. is supported by the Olympia Morata Program of the University of Heidelberg (OM 09/2019). H.P. is supported by the DFG (Projektnummer 360372040 [SFB1335], 395357507 [SFB1371], 324392634 [TRR221]), German Cancer Aid (70114547), the Wilhelm Sander Foundation (2021.040.1), and by the EMBO Young Investigator Program. This study was supported by the generous philanthropic contributions to The University of Texas MD Anderson Cancer Center Moon Shots Program, the Plaform for Innovative Microbiome and Translational Research (PRIME-TR) at the Department of Genomic Medicine at The University of Texas MD Anderson Cancer Center. This work was supported in part by a Cancer Center Support Grant (P30CA016672) from the National Cancer Institute and the Microbiome Core Facility at MD Anderson Cancer Center, and by a Cancer Center Support Grant (P30 CA076292) from the National Cancer Institute to Moffitt Cancer Center and the Moffitt Flow Cytometry Core Facility. R.R.J. is supported by R01HL124112 from the National Institutes of Health. E.E. is supported by the European Research Council, Israel Science Foundation, Israel Ministry of Science and Technology, Israel Ministry of Health, Helmholtz Foundation, Garvan Institute of Medical Research, European Crohn’s and Colitis Organization, Deutsch-Israelische Projektkooperation, IDSA Foundation and Wellcome Trust, and the Charlie Teo Foundation. E.E. is the incumbent of the Sir Marc and Lady Tania Feldmann Professorial Chair, a senior fellow of the Canadian Institute of Advanced Research (CIFAR) and an international scholar of the Bill & Melinda Gates Foundation and Howard Hughes Medical Institute (HHMI).

Author information

Authors and Affiliations

Authors

Contributions

N.Y.S., V.B., M.-L.S., M.A.F., H.P., D.W., M.E., M.H., M.D.J., F.L.L., M.L.D., K.R., M.M., M.H.-V., J.T., E.H., T.H., R.R., L.M., R.G., P.S., R.N., D.C., L.E.F., S.A., S.P.I., R.E.S., P.J., L.J.N., J.W., A.Sc., R.A. and M.L.W. supervised patient recruitment, data and biospecimen collection. R.G., M.D.J., R.R.J., M.Su., C.K.S.-T. and M.D.J., contributed to IRB filing and data safety. N.Y.S., V.B., M.-L.S., H.P., S.S.N., F.L.L., R.E.C., C.R.F., C.M.-T., E.J.S., M.L.D., P.D., M.Sc., M.Su., M.D.J., R.R.J., E.E. and C.K.S.-T. contributed to study concept and design. S.S., R.R., I.F., M.M., M.H.-V. and C.-C.C., performed DNA extraction from stool samples and microbiome sequencing. E.Z., U.M., R.R.J., C.S-T. designed and headed all computational and analytical aspects related to this study. E.Z., U.M., N.Y.S., R.R.J., E.E. and C.K.S.-T. wrote the paper and created the figures and tables. All authors contributed to the final version of the paper. S.S.N., M.Sc., M.Su., M.D.J., R.R.J. and E.E. jointly supervised the work.

Corresponding authors

Correspondence to Neeraj Y. Saini, Michael D. Jain, Robert R. Jenq or Eran Elinav.

Ethics declarations

Competing interests

V.B. received research funding from Bristol Myers-Sqibb (BMS)/Celgene, Gilead, Janssen, Novartis, Roche and Takeda, and honoraria from Gilead, Janssen and Novartis. M.-L.S. is a consultant for Novartis, Gilead and Janssen. H.P. is a consultant for Gilead, Abbvie, Pfizer, Novartis, Servier and BMS, and has received research funding from BMS, and honoraria from Novartis, Gilead, Abbvie, BMS, Servier and Janssen-Cilag. M.L.D. reports consultancy/advisory/honoraria for Kite/Gilead, Novartis, Atara, Precision Biosciences, Celyad, Bellicum, GSK, Adaptive Biotech and Anixa Biosciences, and research funding from Kite/Gilead, Novartis and Atara. F.L.L. reports consultancy/advisory fees from Allogene, Amgen, Bluebird Bio, BMS/Celgene, Calibr, Cellular Biomedicine Group, Cowen, EcoR1, Emerging Therapy Solutions, GammaDelta Therapeutics, Gerson Lehrman Groupt, Iovance, Kite Pharma, Janssen, Legend Biotech, Novartis, Sana, Takeda, Wugen and Umoja, research funding from Kite/Gilead, Allogene, Novartis, BlueBird Bio, BMS, NCI, Leukemia and Lymphoma Society, and education or editorial activity for Aptitude Health, ASH, BioPharma Communications CARE Education, Clinical Care Options Oncology, Imedex and Society for Immunotherapy of Cancer. M.D.J. reports consultancy/advisory fees from Kite/Gilead, Novartis, BMS and MyeloidTx, and research funding from Incyte and Kite/Gilead. R.R.J. has served as a consultant or advisory board member for Merck, Microbiome DX, Karius, MaaT Pharma, LisCure, Seres, Kaleido, and Prolacta and has received patent licence fee or stock options from Seres and Kaleido. N.Y.S., C.-C.C., S.S.N., and R.R.J. are inventors on patent applications by the University of Texas MD Anderson Cancer Center, related to the results of the current study entitled "Serum Metabolomics Related to Chimeric Antigen Receptor (CAR) T-Cell Therapy" and "Gut Microbiome as a Predictive Biomarker of Outcomes for Chimeric Antigen Receptor T-Cell Therapy and its Modulation to Enhance Efficacy and Reduce Toxicity". E.E. is a scientific cofounder of DayTwo and BiomX and an advisor to Hello Inside, Igen and Aposense in topics unrelated to this work. The other authors have no competing interests.

Peer review

Peer review information

Nature Medicine thanks Marcela Maus, Leo Lahti, and Ellen Blaak for their contribution to the peer review of this work. Primary Handling Editor: Joao Monteiro, in collaboration with the Nature Medicine team.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Effects of antibiotic exposure on survival outcomes and on development of toxicities in CD19-targeted CAR-T cell-treated lymphoma patients.

(a) Kaplan–Meier curves for progression-free survival (PFS) in the German and US cohorts displayed separately according to exposure to any antibiotic within 3 weeks before CAR-T cell infusion. (b) Incidence of progression and (c) overall survival in the combined cohort stratified by pre-infusion antibiotic exposure. (d) PFS for the US and the German patient cohorts displayed according to antibiotic risk strata (no antibiotic exposure vs. exposures to low- or to high-risk antibiotics in the three-weeks pre-infusion time window). (e) Histograms of the frequencies of CRS or ICANS grades according to antibiotic risk stratification; statistics by log-rank tests (for survival analyses) and Fisher’s exact tests.

Extended Data Fig. 2 Antibiotic classes administered to CAR-T cell-treated lymphoma patients.

Frequency of individual antibiotic drugs administered to patients within three weeks before (a) and four weeks after (b) CAR-T cell infusion. Histogram of patients receiving individual antibiotics over the course of CAR-T cell therapy for the 8 most frequently prescribed antibiotics in the combined cohort (c) and stratified by country (d). Note that cefepime was only administered to US patients and is therefore not shown in D.

Extended Data Fig. 3 Associations of pre-CAR-T cell infusion antibiotic exposure, tumor burden, performance status, CAR-T cell product and peripheral blood phenotypes.

Serum levels of LDH (a) in German and US patients at the time point of lymphodepletion chemotherapy stratified by whether patients received none or low-risk antibiotics (US: n = 83, Germany: n = 49) versus high-risk antibiotics (US: n = 20: Germany: n = 16). (b) Serum levels of CRP in German and US patients at the time point of lymphodepletion chemotherapy stratified by whether patients received none or low-risk antibiotics versus high-risk antibiotics. (c) IL-6 serum levels in patients at the day of CAR-T cell infusion (none | LR antibiotics: n = 67; HR antibiotics: n = 18). (d) Histograms of the frequencies of bulky lymphomas (>10 cm by radiological measurement), or (e) ECOG performance status according to antibiotic risk stratification. (f) Histograms of the frequencies of antibiotic exposures, or (g) ECOG performance status (left), or ICANS grades (right) according to type of CAR-T cell product administered to the patients. (h) Peripheral blood CD3, CD4 and CD8 T cell counts at the time point of leukapheresis displayed separately for US and German patients. (i) Blood T cell counts stratified by the number of treatment lines prior to CD19-directed CAR-T cell therapy (≤ 3: n = 53; ≥ 4: n = 39). (j and k) FACS analyses of peripheral blood T cell subsets of CAR-T cell-treated patients from Moffitt Cancer Center at the time of leukocyte apheresis and stratified by antibiotic exposure (none | LR antibiotics: n = 39; HR antibiotics: n = 12). CD4 and CD8 stem central memory (SCM; CCR7+, CD45RO-), central memory (CM; CCR7+, CD45RO+), effector memory (EM; CCR7-, CD45RO+), and effector (E; CCR7-CD45RO-) subsets are shown. Statistics for A-C, H, I, K by Mann–Whitney tests, for E-G by Fisher’s exact tests.

Extended Data Fig. 4 Multivariate analysis of antibiotic exposure prior to CD19-targeted CAR-T cell infusion and PFS or OS.

(a) The multivariate Cox model was adjusted for age (stratified by 65 years of age), ECOG (grade 0 and 1 vs. higher), country (Germany vs. US), LDH (normal vs. higher than ULN), CRP (normal vs. higher than 5 mg/dl), bulky disease (> 10 cm) and number of prior therapies (1-3 vs. 4-9). (b) The multivariate Cox model was adjusted for age (stratified by 65 years of age), ECOG (grade 0 and 1 vs. higher), country (Germany vs. US), LDH (normal vs. higher than ULN), CRP (normal vs. higher than 5 mg/dl), bulky disease (> 10 cm) and number of prior therapies (1-3 vs. 4-9). Error bars represent the low and high 95% confidence intervals (CI) of HR; *P < 0.05; **P < 0.01.

Extended Data Fig. 5 Gut microbiome diversity metrics grouped by clinical variables and outcomes.

(a) Number of stool samples collected over the course of CAR-T cell therapy by center and country (n = 351 in total). (b) Shannon indices for alpha diversity of the basal gut microbiome (that is, samples collected between days -21 and 0 relative to CAR-T cell infusion and species composition averaged by mean in case of multiple samples per patient) and grouped for CR vs. no CR at day 180 (left; CR: n = 38, no CR: n = 41 patients), or early progression at day 180 (right; ≤ 180 days: progression within 6 months after infusion (n = 37); no | > 180 days: no progression within follow-up or progression after 6 months after infusion (n = 42 patients) after excluding specimens collected while or less than two weeks after high-risk antibiotics exposure. (c and d) Shannon’s diversity of the basal gut microbiome, after the same exclusion of HR antibiotic samples, grouped for CRS (n = 7, 40, 28, 4 patients [grade 0, 1, 2, 3 + 4]) and ICANS grades (n = 45, 12, 11, 11 patients [grade 0, 1, 2, 3 + 4]). (e) PCoA plots based on Bray–Curtis dissimilarity metrics of the microbiome species and metabolic pathways beta diversity in all samples color-coded for German vs. US patients. (fh) PCoA plots as in (E) for species composition color-coded for ECOG levels (F), number of prior therapy lines (PT; prior to CAR-T cell therapy) (G), and day of specimen collection relative to the day of CAR-T cell infusion (that is, day 0) (H). Group comparisons carried out by Mann–Whitney tests or Kruskal–Wallis rank tests.

Extended Data Fig. 6 Analyzing microbiome features associated with major cohort characteristics.

(a) Differential relative abundance analyses by log-fold changes and statistical testing by Mann–Whitney tests illustrated with volcano plots for species and metabolic pathways encoded in the fecal metagenomes: the log2 abundance in none | LR antibiotic samples (n = 79) / abundance in HR antibiotic samples (n = 16), versus the significance of the abundance difference. Metabolic pathways that have both p-value < 0.05 and absolute log2 fold-change > 1 are shown in red. (b) Bar-plots indicating the fold-change of the top features revealed by the differential abundance analyses described in (A). (c and d) Differential abundance analyses for species and metabolic pathway composition comparing the German (n = 50 patients) and the US (n = 45 patients) cohort. Here, comparisons of fecal metagenomes for samples collected in the 3-weeks pre-CAR-T cell infusion window are shown. Group comparisons carried out by Mann–Whitney tests.

Extended Data Fig. 7 Analyzing microbiome features associated with major outcomes of CAR-T cell therapy.

Differential abundance analyses were done as described in Extended Data Figure 7 with different grouping variables. (a) Left, volcano plots showing species and metabolic pathway compositions of the abundance in patients with survival > 180 day (n = 77) / abundance in patients with survival ≤ 180 day (n = 18) versus the significance of the abundance difference by Mann–Whitney tests. All samples collected in the pre-infusion period were included here. Features with a p-value < 0.05 and absolute log2 change > 1 are shown in red. Middle, bar-plots indicating the fold-change of the top species found in (A). Right, comparison of the relative abundances of the indicated species between patients with survival shorter or longer than 180 days. (b) As in (A), but samples collected during and less than two weeks after exposure to high-risk antibiotics were excluded (alive: n = 66; dead: n = 13). (c and d) As in (B), for metabolic pathways encoded in fecal metagenomes comparing 6-months survival (alive vs. dead at day 180) and early progression (≤180 days: progression within 6 months after infusion vs. no progression within follow-up or progression after day 180; n = 37 vs. n = 42). Group comparisons were computed by Mann–Whitney tests.

Extended Data Fig. 8 Summary of differentially abundant microbiome features by clinical variables and outcomes including all specimens collected in the 3-weeks pre-infusion period.

Heatmaps showing the associations between the species (a) and metabolic pathways (b) of the baseline gut microbiome and clinical parameters and major outcomes (Mann–Whitney tests, p-value < 0.05). Color indicates the Log10 of the following ratios: age (>= 65 / < 65 years), country (Germany / US), ECOG (grades 0-1 / 2-3), LDH (LDH at lymphodepletion chemotherapy: >= 280 / < 280 U ml-1), HR antibiotic exposures (no / yes), response (CR / no CR at day 180), survival (alive / dead at day 180), early progression (lymphoma progression until day 180 / progression > 180 days or no progression during follow-up) and CRS (grades < =1 / >1). Associations with P-value > 0.05 or with absolute log2 fold-change < 1 were set on the heatmap to have a log10 fold-change of zero.

Extended Data Fig. 9 Summary of differentially abundant microbiome features by clinical variables and outcomes excluding baseline specimens collected while or within 2 weeks after exposure to high-risk antibiotics.

Heatmaps showing the associations between the species (a) and metabolic pathways (b) of the baseline gut microbiome and clinical parameters and major outcomes (Mann–Whitney tests, p-value < 0.05). Color indicates the Log10 of the following ratios: age (>= 65 / < 65 years), country (Germany / US), ECOG (grades 0-1 / 2-3), LDH (LDH at lymphodepletion chemotherapy: >= 280 / < 280 U ml-1), response (CR / no CR at day 180), survival (alive / dead at day 180), early progression (lymphoma progression until day 180 / progression > 180 days or no progression during follow-up) and CRS (grades < =1 / >1). Associations with P-value > 0.05 or with absolute log2 fold-change < 1 were set on the heatmap to have a log10 fold-change of zero.

Extended Data Fig. 10 Interaction of antibiotic exposures, survival outcomes and response prediction modeling.

(a) Incidence curves for disease progression in patients were stratified according to high-risk antibiotics exposure in the 3-weeks pre-CAR-T cell infusion time window and according to presence of the peptidoglycan biosynthesis V - pathway in the patients’ stool metagenomes. (b) Deterministic logistic regression models were trained on clinical variables and pre-CAR-T cell infusion gut microbial species composition (mean per patient) of the German cohort and their performance was assessed on the US cohort (with all baseline samples: n = 45 patients; after excluding high-risk antibiotics samples: n = 36 patients). Area under the curve (AUC) receiver operating characteristic (ROC) curves indicating the false-positive and true-positive rates for predicting CR vs. non-CR at day 180 based on an analysis including all patient samples. (c) PCA computed for the whole dataset (that is, training and validation data including high-risk antibiotics samples) based on the features selected during the training process of response at day 180 after CAR-T cell infusion. (d) AUROC curves indicating the false-positive and true-positive rates for predicting CR vs. non-CR at day 180 after training the probabilistic model on the German dataset that excluded high-risk antibiotics exposed samples. (e) AUROC curves indicating the false-positive and true-positive rates for predicting CR vs. non-CR at day 180 based on a deterministic model analysis that excluded samples collected on or less than two weeks after exposure to high-risk antibiotics; data displayed for the US validation cohort. (f) PFS curve of patients after CAR-T cell infusion stratified by the median baseline relative abundance of Bacteroides stercoris as top feature for non-CR as revealed in the machine learning model for CR; only pre-infusion samples without high-risk antibiotic exposure were included. (g) Differences in relative abundances for the four most important species predicting non-response (no CR) grouped by response and high-risk antibiotic exposures; group comparisons carried out by pairwise Mann–Whitney tests with FDR correction of P-values (n = 38 / 41 / 4 / 12 patients [from left to right]); FDR-corrected P-values for B. stercoris are 0.089 (for all three comparisons), for B. fragilis are 0.0725 (for all four comparisons), and for E. sp. CAG38 are 0.0037 (for both comparisons). Statistics in (A) and (F) performed by log-rank tests.

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Stein-Thoeringer, C.K., Saini, N.Y., Zamir, E. et al. A non-antibiotic-disrupted gut microbiome is associated with clinical responses to CD19-CAR-T cell cancer immunotherapy. Nat Med 29, 906–916 (2023). https://doi.org/10.1038/s41591-023-02234-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-023-02234-6

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer