Advertisement
No access
Report

Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors

Bertrand Routy https://orcid.org/0000-0002-0756-6571, Emmanuelle Le Chatelier https://orcid.org/0000-0002-2724-0536, Lisa Derosa, Connie P. M. Duong https://orcid.org/0000-0002-9067-8557, Maryam Tidjani Alou, Romain Daillère https://orcid.org/0000-0003-3617-6377, Aurélie Fluckiger https://orcid.org/0000-0001-7135-0049, Meriem Messaoudene, Conrad Rauber, Maria P. Roberti, Marine Fidelle, Caroline Flament, Vichnou Poirier-Colame, Paule Opolon, Christophe Klein, Kristina Iribarren https://orcid.org/0000-0002-2930-8573, Laura Mondragón https://orcid.org/0000-0002-3257-045X, Nicolas Jacquelot https://orcid.org/0000-0003-0282-1892, Bo Qu https://orcid.org/0000-0001-7947-3350, Gladys Ferrere, Céline Clémenson, Laura Mezquita, Jordi Remon Masip https://orcid.org/0000-0001-9839-149X, Charles Naltet, Solenn Brosseau, Coureche Kaderbhai, Corentin Richard, Hira Rizvi, Florence Levenez, Nathalie Galleron, Benoit Quinquis, Nicolas Pons https://orcid.org/0000-0003-1926-8077, Bernhard Ryffel, Véronique Minard-Colin, Patrick Gonin https://orcid.org/0000-0001-6151-4580, Jean-Charles Soria, Eric Deutsch https://orcid.org/0000-0002-8223-3697, Yohann Loriot, François Ghiringhelli, Gérard Zalcman https://orcid.org/0000-0002-0343-9575, François Goldwasser, Bernard Escudier https://orcid.org/0000-0002-4626-0479, Matthew D. Hellmann https://orcid.org/0000-0002-2670-9777, Alexander Eggermont https://orcid.org/0000-0001-7458-4326, Didier Raoult, Laurence Albiges, Guido Kroemer https://orcid.org/0000-0002-9334-4405 [email protected], and Laurence Zitvogel https://orcid.org/0000-0003-1596-0998 [email protected]Authors Info & Affiliations
Science
2 Nov 2017
Vol 359, Issue 6371
pp. 91-97

Good bacteria help fight cancer

Resident gut bacteria can affect patient responses to cancer immunotherapy (see the Perspective by Jobin). Routy et al. show that antibiotic consumption is associated with poor response to immunotherapeutic PD-1 blockade. They profiled samples from patients with lung and kidney cancers and found that nonresponding patients had low levels of the bacterium Akkermansia muciniphila. Oral supplementation of the bacteria to antibiotic-treated mice restored the response to immunotherapy. Matson et al. and Gopalakrishnan et al. studied melanoma patients receiving PD-1 blockade and found a greater abundance of “good” bacteria in the guts of responding patients. Nonresponders had an imbalance in gut flora composition, which correlated with impaired immune cell activity. Thus, maintaining healthy gut flora could help patients combat cancer.
Science, this issue p. 91, p. 104, p. 97; see also p. 32

Abstract

Immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 axis induce sustained clinical responses in a sizable minority of cancer patients. We found that primary resistance to ICIs can be attributed to abnormal gut microbiome composition. Antibiotics inhibited the clinical benefit of ICIs in patients with advanced cancer. Fecal microbiota transplantation (FMT) from cancer patients who responded to ICIs into germ-free or antibiotic-treated mice ameliorated the antitumor effects of PD-1 blockade, whereas FMT from nonresponding patients failed to do so. Metagenomics of patient stool samples at diagnosis revealed correlations between clinical responses to ICIs and the relative abundance of Akkermansia muciniphila. Oral supplementation with A. muciniphila after FMT with nonresponder feces restored the efficacy of PD-1 blockade in an interleukin-12–dependent manner by increasing the recruitment of CCR9+CXCR3+CD4+ T lymphocytes into mouse tumor beds.

Get full access to this article

View all available purchase options and get full access to this article.

Supplementary Material

Summary

Materials and Methods
Figs. S1 to S16
Tables S1 to S18
References (3135)

Resources

File (aan3706_routy_sm.pdf)

References and Notes

1
C. Robert, L. Thomas, I. Bondarenko, S. O’Day, J. Weber, C. Garbe, C. Lebbe, J.-F. Baurain, A. Testori, J.-J. Grob, N. Davidson, J. Richards, M. Maio, A. Hauschild, W. H. Miller Jr., P. Gascon, M. Lotem, K. Harmankaya, R. Ibrahim, S. Francis, T.-T. Chen, R. Humphrey, A. Hoos, J. D. Wolchok, Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N. Engl. J. Med. 364, 2517–2526 (2011).
2
N. P. Restifo, M. E. Dudley, S. A. Rosenberg, Adoptive immunotherapy for cancer: Harnessing the T cell response. Nat. Rev. Immunol. 12, 269–281 (2012).
3
H. Borghaei, L. Paz-Ares, L. Horn, D. R. Spigel, M. Steins, N. E. Ready, L. Q. Chow, E. E. Vokes, E. Felip, E. Holgado, F. Barlesi, M. Kohlhäufl, O. Arrieta, M. A. Burgio, J. Fayette, H. Lena, E. Poddubskaya, D. E. Gerber, S. N. Gettinger, C. M. Rudin, N. Rizvi, L. Crinò, G. R. Blumenschein Jr., S. J. Antonia, C. Dorange, C. T. Harbison, F. Graf Finckenstein, J. R. Brahmer, Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N. Engl. J. Med. 373, 1627–1639 (2015).
4
S. M. Ansell, A. M. Lesokhin, I. Borrello, A. Halwani, E. C. Scott, M. Gutierrez, S. J. Schuster, M. M. Millenson, D. Cattry, G. J. Freeman, S. J. Rodig, B. Chapuy, A. H. Ligon, L. Zhu, J. F. Grosso, S. Y. Kim, J. M. Timmerman, M. A. Shipp, P. Armand, PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N. Engl. J. Med. 372, 311–319 (2015).
5
S. L. Topalian, F. S. Hodi, J. R. Brahmer, S. N. Gettinger, D. C. Smith, D. F. McDermott, J. D. Powderly, R. D. Carvajal, J. A. Sosman, M. B. Atkins, P. D. Leming, D. R. Spigel, S. J. Antonia, L. Horn, C. G. Drake, D. M. Pardoll, L. Chen, W. H. Sharfman, R. A. Anders, J. M. Taube, T. L. McMiller, H. Xu, A. J. Korman, M. Jure-Kunkel, S. Agrawal, D. McDonald, G. D. Kollia, A. Gupta, J. M. Wigginton, M. Sznol, Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366, 2443–2454 (2012).
6
A. Ribas, O. Hamid, A. Daud, F. S. Hodi, J. D. Wolchok, R. Kefford, A. M. Joshua, A. Patnaik, W.-J. Hwu, J. S. Weber, T. C. Gangadhar, P. Hersey, R. Dronca, R. W. Joseph, H. Zarour, B. Chmielowski, D. P. Lawrence, A. Algazi, N. A. Rizvi, B. Hoffner, C. Mateus, K. Gergich, J. A. Lindia, M. Giannotti, X. N. Li, S. Ebbinghaus, S. P. Kang, C. Robert, Association of pembrolizumab with tumor response and survival among patients with advanced melanoma. J. Am. Med. Assoc. 315, 1600–1609 (2016).
7
D. Pardoll, Cancer and the immune system: Basic concepts and targets for intervention. Semin. Oncol. 42, 523–538 (2015).
8
R. J. Motzer, B. Escudier, D. F. McDermott, S. George, H. J. Hammers, S. Srinivas, S. S. Tykodi, J. A. Sosman, G. Procopio, E. R. Plimack, D. Castellano, T. K. Choueiri, H. Gurney, F. Donskov, P. Bono, J. Wagstaff, T. C. Gauler, T. Ueda, Y. Tomita, F. A. Schutz, C. Kollmannsberger, J. Larkin, A. Ravaud, J. S. Simon, L.-A. Xu, I. M. Waxman, P. Sharma, Nivolumab versus everolimus in advanced renal-cell carcinoma. N. Engl. J. Med. 373, 1803–1813 (2015).
9
N. A. Rizvi, M. D. Hellmann, A. Snyder, P. Kvistborg, V. Makarov, J. J. Havel, W. Lee, J. Yuan, P. Wong, T. S. Ho, M. L. Miller, N. Rekhtman, A. L. Moreira, F. Ibrahim, C. Bruggeman, B. Gasmi, R. Zappasodi, Y. Maeda, C. Sander, E. B. Garon, T. Merghoub, J. D. Wolchok, T. N. Schumacher, T. A. Chan, Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer. Science 348, 124–128 (2015).
10
N. Riaz, J. J. Havel, S. M. Kendall, V. Makarov, L. A. Walsh, A. Desrichard, N. Weinhold, T. A. Chan, Recurrent SERPINB3 and SERPINB4 mutations in patients who respond to anti-CTLA4 immunotherapy. Nat. Genet. 48, 1327–1329 (2016).
11
D. P. Carbone, M. Reck, L. Paz-Ares, B. Creelan, L. Horn, M. Steins, E. Felip, M. M. van den Heuvel, T.-E. Ciuleanu, F. Badin, N. Ready, T. J. N. Hiltermann, S. Nair, R. Juergens, S. Peters, E. Minenza, J. M. Wrangle, D. Rodriguez-Abreu, H. Borghaei, G. R. Blumenschein Jr., L. C. Villaruz, L. Havel, J. Krejci, J. Corral Jaime, H. Chang, W. J. Geese, P. Bhagavatheeswaran, A. C. Chen, M. A. Socinski, First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N. Engl. J. Med. 376, 2415–2426 (2017).
12
T. N. Schumacher, R. D. Schreiber, Neoantigens in cancer immunotherapy. Science 348, 69–74 (2015).
13
S. Spranger, R. Bao, T. F. Gajewski, Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 523, 231–235 (2015).
14
M. J. Smyth, S. F. Ngiow, A. Ribas, M. W. L. Teng, Combination cancer immunotherapies tailored to the tumour microenvironment. Nat. Rev. Clin. Oncol. 13, 143–158 (2016).
15
S. Koyama, E. A. Akbay, Y. Y. Li, G. S. Herter-Sprie, K. A. Buczkowski, W. G. Richards, L. Gandhi, A. J. Redig, S. J. Rodig, H. Asahina, R. E. Jones, M. M. Kulkarni, M. Kuraguchi, S. Palakurthi, P. E. Fecci, B. E. Johnson, P. A. Janne, J. A. Engelman, S. P. Gangadharan, D. B. Costa, G. J. Freeman, R. Bueno, F. S. Hodi, G. Dranoff, K.-K. Wong, P. S. Hammerman, Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat. Commun. 7, 10501 (2016).
16
J. U. Peled, S. M. Devlin, A. Staffas, M. Lumish, R. Khanin, E. R. Littmann, L. Ling, S. Kosuri, M. Maloy, J. B. Slingerland, K. F. Ahr, K. A. Porosnicu Rodriguez, Y. Shono, A. E. Slingerland, M. D. Docampo, A. D. Sung, D. Weber, A. M. Alousi, B. Gyurkocza, D. M. Ponce, J. N. Barker, M.-A. Perales, S. A. Giralt, Y. Taur, E. G. Pamer, R. R. Jenq, M. R. M. van den Brink, Intestinal microbiota and relapse after hematopoietic-cell transplantation. J. Clin. Oncol. 35, 1650–1659 (2017).
17
S. Viaud, F. Saccheri, G. Mignot, T. Yamazaki, R. Daillère, D. Hannani, D. P. Enot, C. Pfirschke, C. Engblom, M. J. Pittet, A. Schlitzer, F. Ginhoux, L. Apetoh, E. Chachaty, P.-L. Woerther, G. Eberl, M. Bérard, C. Ecobichon, D. Clermont, C. Bizet, V. Gaboriau-Routhiau, N. Cerf-Bensussan, P. Opolon, N. Yessaad, E. Vivier, B. Ryffel, C. O. Elson, J. Doré, G. Kroemer, P. Lepage, I. G. Boneca, F. Ghiringhelli, L. Zitvogel, The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science 342, 971–976 (2013).
18
N. Iida, A. Dzutsev, C. A. Stewart, L. Smith, N. Bouladoux, R. A. Weingarten, D. A. Molina, R. Salcedo, T. Back, S. Cramer, R.-M. Dai, H. Kiu, M. Cardone, S. Naik, A. K. Patri, E. Wang, F. M. Marincola, K. M. Frank, Y. Belkaid, G. Trinchieri, R. S. Goldszmid, Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 342, 967–970 (2013).
19
R. Daillère, M. Vétizou, N. Waldschmitt, T. Yamazaki, C. Isnard, V. Poirier-Colame, C. P. M. Duong, C. Flament, P. Lepage, M. P. Roberti, B. Routy, N. Jacquelot, L. Apetoh, S. Becharef, S. Rusakiewicz, P. Langella, H. Sokol, G. Kroemer, D. Enot, A. Roux, A. Eggermont, E. Tartour, L. Johannes, P.-L. Woerther, E. Chachaty, J.-C. Soria, E. Golden, S. Formenti, M. Plebanski, M. Madondo, P. Rosenstiel, D. Raoult, V. Cattoir, I. G. Boneca, M. Chamaillard, L. Zitvogel, Enterococcus hirae and Barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects. Immunity 45, 931–943 (2016).
20
M. Vétizou, J. M. Pitt, R. Daillère, P. Lepage, N. Waldschmitt, C. Flament, S. Rusakiewicz, B. Routy, M. P. Roberti, C. P. M. Duong, V. Poirier-Colame, A. Roux, S. Becharef, S. Formenti, E. Golden, S. Cording, G. Eberl, A. Schlitzer, F. Ginhoux, S. Mani, T. Yamazaki, N. Jacquelot, D. P. Enot, M. Bérard, J. Nigou, P. Opolon, A. Eggermont, P.-L. Woerther, E. Chachaty, N. Chaput, C. Robert, C. Mateus, G. Kroemer, D. Raoult, I. G. Boneca, F. Carbonnel, M. Chamaillard, L. Zitvogel, Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 350, 1079–1084 (2015).
21
A. Sivan, L. Corrales, N. Hubert, J. B. Williams, K. Aquino-Michaels, Z. M. Earley, F. W. Benyamin, Y. M. Lei, B. Jabri, M.-L. Alegre, E. B. Chang, T. F. Gajewski, Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 350, 1084–1089 (2015).
22
M. A. Jackson, J. K. Goodrich, M.-E. Maxan, D. E. Freedberg, J. A. Abrams, A. C. Poole, J. L. Sutter, D. Welter, R. E. Ley, J. T. Bell, T. D. Spector, C. J. Steves, Proton pump inhibitors alter the composition of the gut microbiota. Gut 65, 749–756 (2016).
23
M. J. Blaser, Antibiotic use and its consequences for the normal microbiome. Science 352, 544–545 (2016).
24
J. Li, H. Jia, X. Cai, H. Zhong, Q. Feng, S. Sunagawa, M. Arumugam, J. R. Kultima, E. Prifti, T. Nielsen, A. S. Juncker, C. Manichanh, B. Chen, W. Zhang, F. Levenez, J. Wang, X. Xu, L. Xiao, S. Liang, D. Zhang, Z. Zhang, W. Chen, H. Zhao, J. Y. Al-Aama, S. Edris, H. Yang, J. Wang, T. Hansen, H. B. Nielsen, S. Brunak, K. Kristiansen, F. Guarner, O. Pedersen, J. Doré, S. D. Ehrlich, P. Bork, J. Wang, J.-M. Batto, S. Kennedy, F. Haimet, Y. Winogradski, E. Pelletier, D. LePaslier, F. Artiguenave, T. Bruls, J. Weissenbach, K. Turner, J. Parkhill, M. Antolin, F. Casellas, N. Borruel, E. Varela, A. Torrejon, G. Denariaz, M. Derrien, J. E. T. van Hylckama Vlieg, P. Viega, R. Oozeer, J. Knoll, M. Rescigno, C. Brechot, C. M’Rini, A. Mérieux, T. Yamada, S. Tims, E. G. Zoetendal, M. Kleerebezem, W. M. de Vos, A. Cultrone, M. Leclerc, C. Juste, E. Guedon, C. Delorme, S. Layec, G. Khaci, M. van de Guchte, G. Vandemeulebrouck, A. Jamet, R. Dervyn, N. Sanchez, H. Blottière, E. Maguin, P. Renault, J. Tap, D. R. Mende, An integrated catalog of reference genes in the human gut microbiome. Nat. Biotechnol. 32, 834–841 (2014).
25
E. A. Eisenhauer, P. Therasse, J. Bogaerts, L. H. Schwartz, D. Sargent, R. Ford, J. Dancey, S. Arbuck, S. Gwyther, M. Mooney, L. Rubinstein, L. Shankar, L. Dodd, R. Kaplan, D. Lacombe, J. Verweij, New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). Eur. J. Cancer 45, 228–247 (2009).
26
X. Wang, J. E. Schoenhals, A. Li, D. R. Valdecanas, H. Ye, F. Zang, C. Tang, M. Tang, C.-G. Liu, X. Liu, S. Krishnan, J. P. Allison, P. Sharma, P. Hwu, R. Komaki, W. W. Overwijk, D. R. Gomez, J. Y. Chang, S. M. Hahn, M. A. Cortez, J. W. Welsh, Suppression of type I IFN signaling in tumors mediates resistance to anti-PD-1 treatment that can be overcome by radiotherapy. Cancer Res. 77, 839–850 (2017).
27
M. C. Collado, M. Derrien, E. Isolauri, W. M. de Vos, S. Salminen, Intestinal integrity and Akkermansia muciniphila, a mucin-degrading member of the intestinal microbiota present in infants, adults, and the elderly. Appl. Environ. Microbiol. 73, 7767–7770 (2007).
28
N. Geva-Zatorsky, E. Sefik, L. Kua, L. Pasman, T. G. Tan, A. Ortiz-Lopez, T. B. Yanortsang, L. Yang, R. Jupp, D. Mathis, C. Benoist, D. L. Kasper, Mining the human gut microbiota for immunomodulatory organisms. Cell 168, 928–943.e11 (2017).
29
A. Suau, R. Bonnet, M. Sutren, J. J. Godon, G. R. Gibson, M. D. Collins, J. Doré, Direct analysis of genes encoding 16S rRNA from complex communities reveals many novel molecular species within the human gut. Appl. Environ. Microbiol. 65, 4799–4807 (1999).
30
D. S. Chen, I. Mellman, Elements of cancer immunity and the cancer-immune set point. Nature 541, 321–330 (2017).
31
A. Criscuolo, S. Brisse, AlienTrimmer: A tool to quickly and accurately trim off multiple short contaminant sequences from high-throughput sequencing reads. Genomics 102, 500–506 (2013).
32
B. Langmead, S. L. Salzberg, Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).
33
A. Cotillard, S. P. Kennedy, L. C. Kong, E. Prifti, N. Pons, E. Le Chatelier, M. Almeida, B. Quinquis, F. Levenez, N. Galleron, S. Gougis, S. Rizkalla, J.-M. Batto, P. Renault, J. Doré, J. D. Zucker, K. Clément, S. D. Ehrlich, S. D. Ehrlich, H. Blottière, M. Leclerc, C. Juste, T. de Wouters, P. Lepage, C. Fouqueray, A. Basdevant, C. Henegar, C. Godard, M. Fondacci, A. Rohia, F. Hajduch, J. Weissenbach, E. Pelletier, D. Le Paslier, J.-P. Gauchi, J.-F. Gibrat, V. Loux, W. Carré, E. Maguin, M. van de Guchte, A. Jamet, F. Boumezbeur, S. Layec, Dietary intervention impact on gut microbial gene richness. Nature 500, 585–588 (2013).
34
E. Le Chatelier, T. Nielsen, J. Qin, E. Prifti, F. Hildebrand, G. Falony, M. Almeida, M. Arumugam, J.-M. Batto, S. Kennedy, P. Leonard, J. Li, K. Burgdorf, N. Grarup, T. Jørgensen, I. Brandslund, H. B. Nielsen, A. S. Juncker, M. Bertalan, F. Levenez, N. Pons, S. Rasmussen, S. Sunagawa, J. Tap, S. Tims, E. G. Zoetendal, S. Brunak, K. Clément, J. Doré, M. Kleerebezem, K. Kristiansen, P. Renault, T. Sicheritz-Ponten, W. M. de Vos, J.-D. Zucker, J. Raes, T. Hansen, P. Bork, J. Wang, S. D. Ehrlich, O. Pedersen, E. Guedon, C. Delorme, S. Layec, G. Khaci, M. van de Guchte, G. Vandemeulebrouck, A. Jamet, R. Dervyn, N. Sanchez, E. Maguin, F. Haimet, Y. Winogradski, A. Cultrone, M. Leclerc, C. Juste, H. Blottière, E. Pelletier, D. LePaslier, F. Artiguenave, T. Bruls, J. Weissenbach, K. Turner, J. Parkhill, M. Antolin, C. Manichanh, F. Casellas, N. Boruel, E. Varela, A. Torrejon, F. Guarner, G. Denariaz, M. Derrien, J. E. T. van Hylckama Vlieg, P. Veiga, R. Oozeer, J. Knol, M. Rescigno, C. Brechot, C. M’Rini, A. Mérieux, T. Yamada, Richness of human gut microbiome correlates with metabolic markers. Nature 500, 541–546 (2013).
35
H. B. Nielsen, M. Almeida, A. S. Juncker, S. Rasmussen, J. Li, S. Sunagawa, D. R. Plichta, L. Gautier, A. G. Pedersen, E. Le Chatelier, E. Pelletier, I. Bonde, T. Nielsen, C. Manichanh, M. Arumugam, J.-M. Batto, M. B. Quintanilha Dos Santos, N. Blom, N. Borruel, K. S. Burgdorf, F. Boumezbeur, F. Casellas, J. Doré, P. Dworzynski, F. Guarner, T. Hansen, F. Hildebrand, R. S. Kaas, S. Kennedy, K. Kristiansen, J. R. Kultima, P. Léonard, F. Levenez, O. Lund, B. Moumen, D. Le Paslier, N. Pons, O. Pedersen, E. Prifti, J. Qin, J. Raes, S. Sørensen, J. Tap, S. Tims, D. W. Ussery, T. Yamada, P. Renault, T. Sicheritz-Ponten, P. Bork, J. Wang, S. Brunak, S. D. Ehrlich, A. Jamet, A. Mérieux, A. Cultrone, A. Torrejon, B. Quinquis, C. Brechot, C. Delorme, C. M’Rini, W. M. de Vos, E. Maguin, E. Varela, E. Guedon, F. Gwen, F. Haimet, F. Artiguenave, G. Vandemeulebrouck, G. Denariaz, G. Khaci, H. Blottière, J. Knol, J. Weissenbach, J. E. T. van Hylckama Vlieg, J. Torben, J. Parkhill, K. Turner, M. van de Guchte, M. Antolin, M. Rescigno, M. Kleerebezem, M. Derrien, N. Galleron, N. Sanchez, N. Grarup, P. Veiga, R. Oozeer, R. Dervyn, S. Layec, T. Bruls, Y. Winogradski, E. G. Zoetendal, Identification and assembly of genomes and genetic elements in complex metagenomic samples without using reference genomes. Nat. Biotechnol. 32, 822–828 (2014).

(0)eLetters

eLetters is a forum for ongoing peer review. eLetters are not edited, proofread, or indexed, but they are screened. eLetters should provide substantive and scholarly commentary on the article. Embedded figures cannot be submitted, and we discourage the use of figures within eLetters in general. If a figure is essential, please include a link to the figure within the text of the eLetter. Please read our Terms of Service before submitting an eLetter.

Log In to Submit a Response

No eLetters have been published for this article yet.

Information & Authors

Information

Published In

Science
Volume 359 | Issue 6371
5 January 2018

Article versions

You are viewing the most recent version of this article.

Submission history

Received: 5 April 2017
Accepted: 20 September 2017
Published in print: 5 January 2018

Permissions

Request permissions for this article.

Acknowledgments

We thank the animal facility team and V. Rouffiac from the imaging platform of Gustave Roussy, as well as S. Jabs for technical help. The data reported in this manuscript are tabulated in the main paper and the supplementary materials. The metagenomic shotgun sequencing data are available from the European Nucleotide Archive (EMBL-EBI) under accession number PRJEB22863. B.R. was supported by Gustave Roussy Course of Excellence in Oncology–Fondation Philanthropia and a McGill University Townsend hematology research fellowship award. L.Z. and G.K. were supported by the Ligue contre le Cancer (équipe labelisée), Agence Nationale de la Recherche (ANR)–Projets blancs, ANR under the frame of E-Rare-2 (ERA-Net for Research on Rare Diseases), Association pour la recherche sur le cancer (ARC), Cancéropôle Ile-de-France, Institut National du Cancer (INCa), Institut Universitaire de France, Fondation pour la Recherche Médicale (FRM), the European Commission (ArtForce), the European Research Council (ERC), the LeDucq Foundation, LabEx Immuno-Oncology, SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination (SOCRATE), SIRIC Cancer Research and Personalized Medicine (CARPEM), the Paris Alliance of Cancer Research Institutes (PACRI), and philanthropia (E. Badinter and N. Meyer). L.Z., E.L.C., and B.R. are inventors on patent EP17305206 and European license 16306779.6 held by Institut Gustave Roussy that covers use of microbial modulators for PD1/PD-L1/PD-L2 mAb-based treatments. L.Z. and G.K. are cofounders of EverImmune, a biotech company focused on the use of commensal bacteria for cancer treatment.

Authors

Affiliations

Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Institut National de la Santé et de la Recherche Medicale (INSERM) U1015 and Equipe Labellisée–Ligue Nationale contre le Cancer, Villejuif, France.
Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France.
Emmanuelle Le Chatelier https://orcid.org/0000-0002-2724-0536
MGP MetaGénoPolis, INRA, Université Paris-Saclay, Jouy-en-Josas, France.
Lisa Derosa
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Institut National de la Santé et de la Recherche Medicale (INSERM) U1015 and Equipe Labellisée–Ligue Nationale contre le Cancer, Villejuif, France.
Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France.
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Institut National de la Santé et de la Recherche Medicale (INSERM) U1015 and Equipe Labellisée–Ligue Nationale contre le Cancer, Villejuif, France.
Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.
Maryam Tidjani Alou
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Institut National de la Santé et de la Recherche Medicale (INSERM) U1015 and Equipe Labellisée–Ligue Nationale contre le Cancer, Villejuif, France.
Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France.
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Institut National de la Santé et de la Recherche Medicale (INSERM) U1015 and Equipe Labellisée–Ligue Nationale contre le Cancer, Villejuif, France.
Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France.
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Institut National de la Santé et de la Recherche Medicale (INSERM) U1015 and Equipe Labellisée–Ligue Nationale contre le Cancer, Villejuif, France.
Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.
Meriem Messaoudene
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Institut National de la Santé et de la Recherche Medicale (INSERM) U1015 and Equipe Labellisée–Ligue Nationale contre le Cancer, Villejuif, France.
Conrad Rauber
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Institut National de la Santé et de la Recherche Medicale (INSERM) U1015 and Equipe Labellisée–Ligue Nationale contre le Cancer, Villejuif, France.
Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France.
Maria P. Roberti
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Institut National de la Santé et de la Recherche Medicale (INSERM) U1015 and Equipe Labellisée–Ligue Nationale contre le Cancer, Villejuif, France.
Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.
Marine Fidelle
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France.
Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.
Caroline Flament
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Institut National de la Santé et de la Recherche Medicale (INSERM) U1015 and Equipe Labellisée–Ligue Nationale contre le Cancer, Villejuif, France.
Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.
Vichnou Poirier-Colame
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Institut National de la Santé et de la Recherche Medicale (INSERM) U1015 and Equipe Labellisée–Ligue Nationale contre le Cancer, Villejuif, France.
Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.
Paule Opolon
Gustave Roussy, Laboratoire de Pathologie Expérimentale, 94800 Villejuif, France.
Christophe Klein
Centre de Recherche des Cordeliers, INSERM, Université Paris Descartes, Sorbonne Paris Cité, UMRS 1138, Université Pierre et Marie Curie Université Paris 06, Sorbonne Universités, Paris, France.
Metabolomics and Cell Biology Platforms, GRCC, Villejuif, France.
Paris Descartes University, Sorbonne Paris Cité, Paris, France.
Equipe 11 Labellisée–Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.
INSERM U1138, Paris, France.
Université Pierre et Marie Curie, Paris, France.
Metabolomics and Cell Biology Platforms, GRCC, Villejuif, France.
Paris Descartes University, Sorbonne Paris Cité, Paris, France.
Equipe 11 Labellisée–Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.
INSERM U1138, Paris, France.
Université Pierre et Marie Curie, Paris, France.
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Institut National de la Santé et de la Recherche Medicale (INSERM) U1015 and Equipe Labellisée–Ligue Nationale contre le Cancer, Villejuif, France.
Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France.
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Institut National de la Santé et de la Recherche Medicale (INSERM) U1015 and Equipe Labellisée–Ligue Nationale contre le Cancer, Villejuif, France.
Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France.
Gladys Ferrere
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Institut National de la Santé et de la Recherche Medicale (INSERM) U1015 and Equipe Labellisée–Ligue Nationale contre le Cancer, Villejuif, France.
Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France.
Céline Clémenson
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Department of Radiation Oncology, INSERM U1030, and Molecular Radiotherapy, Gustave Roussy, Université Paris-Saclay, F-94805 Villejuif, France.
Laura Mezquita
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Department of Medical Oncology, Gustave Roussy, Villejuif, France.
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Department of Medical Oncology, Gustave Roussy, Villejuif, France.
Charles Naltet
Thoracic Oncology Department–CIC1425/CLIP2 Paris-Nord, Hospital Bichat-Claude Bernard, AP-HP, Université Paris-Diderot, Paris, France.
Solenn Brosseau
Thoracic Oncology Department–CIC1425/CLIP2 Paris-Nord, Hospital Bichat-Claude Bernard, AP-HP, Université Paris-Diderot, Paris, France.
Coureche Kaderbhai
Department of Medical Oncology, Center GF Leclerc, Dijon, France.
Corentin Richard
Department of Medical Oncology, Center GF Leclerc, Dijon, France.
Hira Rizvi
Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
Florence Levenez
MGP MetaGénoPolis, INRA, Université Paris-Saclay, Jouy-en-Josas, France.
Nathalie Galleron
MGP MetaGénoPolis, INRA, Université Paris-Saclay, Jouy-en-Josas, France.
Benoit Quinquis
MGP MetaGénoPolis, INRA, Université Paris-Saclay, Jouy-en-Josas, France.
MGP MetaGénoPolis, INRA, Université Paris-Saclay, Jouy-en-Josas, France.
Bernhard Ryffel
Molecular Immunology and Embryology, UMR 7355, CNRS, University of Orleans, Orléans, France.
Véronique Minard-Colin
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Department of Pediatric Oncology, GRCC, Villejuif, France.
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Preclinical Research Platform, GRCC, Villejuif, France.
Jean-Charles Soria
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Department of Medical Oncology, Gustave Roussy, Villejuif, France.
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Department of Radiation Oncology, INSERM U1030, and Molecular Radiotherapy, Gustave Roussy, Université Paris-Saclay, F-94805 Villejuif, France.
Yohann Loriot
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France.
Department of Medical Oncology, Gustave Roussy, Villejuif, France.
François Ghiringhelli
Department of Medical Oncology, Center GF Leclerc, Dijon, France.
Thoracic Oncology Department–CIC1425/CLIP2 Paris-Nord, Hospital Bichat-Claude Bernard, AP-HP, Université Paris-Diderot, Paris, France.
François Goldwasser
Paris Descartes University, Sorbonne Paris Cité, Paris, France.
Department of Medical Oncology, Cochin Hospital, Assistance Publique–Hôpitaux de Paris, Paris, France.
Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Paris, France.
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Department of Medical Oncology, Gustave Roussy, Villejuif, France.
INSERM U981, GRCC, Villejuif, France.
Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Institut National de la Santé et de la Recherche Medicale (INSERM) U1015 and Equipe Labellisée–Ligue Nationale contre le Cancer, Villejuif, France.
Department of Medical Oncology, Gustave Roussy, Villejuif, France.
Didier Raoult
URMITE, Aix Marseille Université, UM63, CNRS 7278, IRD 198, INSERM 1095, IHU–Méditerranée Infection, 13005 Marseille, France.
Laurence Albiges
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France.
Department of Medical Oncology, Gustave Roussy, Villejuif, France.
Metabolomics and Cell Biology Platforms, GRCC, Villejuif, France.
Paris Descartes University, Sorbonne Paris Cité, Paris, France.
Equipe 11 Labellisée–Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.
INSERM U1138, Paris, France.
Université Pierre et Marie Curie, Paris, France.
Pôle de Biologie, Hôpital Européen Georges Pompidou, Assistance Publique–Hôpitaux de Paris, Paris, France.
Department of Women’s and Children’s Health, Karolinska University Hospital, 17176 Stockholm, Sweden.
Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
Institut National de la Santé et de la Recherche Medicale (INSERM) U1015 and Equipe Labellisée–Ligue Nationale contre le Cancer, Villejuif, France.
Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France.
Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.

Notes

*
Corresponding author. Email: [email protected] (L.Z.); [email protected] (G.K.)

Metrics & Citations

Metrics

Article Usage

Altmetrics

Citations

Cite as

Export citation

Select the format you want to export the citation of this publication.

Cited by

  1. Microbes and vitamin D aid immunotherapy, Science, 384, 6694, (384-385), (2024)./doi/10.1126/science.adp1309
    Abstract
  2. Vitamin D regulates microbiome-dependent cancer immunity, Science, 384, 6694, (428-437), (2024)./doi/10.1126/science.adh7954
    Abstract
  3. Current Advances in Immune Checkpoint Therapy, Immune Checkpoint Inhibitors - New Insights and Recent Progress, (2023).https://doi.org/10.5772/intechopen.107315
    Crossref
  4. Genetic heterogeneity of colorectal cancer and the microbiome, World Journal of Gastrointestinal Oncology, 15, 3, (443-463), (2023).https://doi.org/10.4251/wjgo.v15.i3.443
    Crossref
  5. Challenges and exploration for immunotherapies targeting cold colorectal cancer, World Journal of Gastrointestinal Oncology, 15, 1, (55-68), (2023).https://doi.org/10.4251/wjgo.v15.i1.55
    Crossref
  6. Microbial Components and Effector Molecules in T Helper Cell Differentiation and Function, Immune Network, 23, 1, (2023).https://doi.org/10.4110/in.2023.23.e7
    Crossref
  7. Molecular mechanisms targeting drug-resistance and metastasis in colorectal cancer: Updates and beyond, World Journal of Gastroenterology, 29, 9, (1395-1426), (2023).https://doi.org/10.3748/wjg.v29.i9.1395
    Crossref
  8. Eubacterium rectale Improves the Efficacy of Anti-PD1 Immunotherapy in Melanoma via l-Serine-Mediated NK Cell Activation, Research, 6, (2023)./doi/10.34133/research.0127
    Abstract
  9. Health and Disease: Akkermansia muciniphila, the Shining Star of the Gut Flora, Research, 6, (2023)./doi/10.34133/research.0107
    Abstract
  10. Potential Role of Vaginal Microbiota in Ovarian Cancer Carcinogenesis, Progression and Treatment, Pharmaceutics, 15, 3, (948), (2023).https://doi.org/10.3390/pharmaceutics15030948
    Crossref
  11. See more
Loading...

View Options

Check Access

Log in to view the full text

AAAS ID LOGIN

AAAS login provides access to Science for AAAS Members, and access to other journals in the Science family to users who have purchased individual subscriptions.

Log in via OpenAthens.
Log in via Shibboleth.

More options

Register for free to read this article

As a service to the community, this article is available for free. Login or register for free to read this article.

Purchase this issue in print

Buy a single issue of Science for just $15 USD.

View options

PDF format

Download this article as a PDF file

Download PDF

Full Text

FULL TEXT

Media

Figures

Multimedia

Tables

Share

Share

Share article link

Share on social media