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:

Anti-TIGIT antibody improves PD-L1 blockade through myeloid and Treg cells

A Publisher Correction to this article was published on 13 March 2024

This article has been updated

Abstract

Tiragolumab, an anti-TIGIT antibody with an active IgG1κ Fc, demonstrated improved outcomes in the phase 2 CITYSCAPE trial (ClinicalTrials.gov: NCT03563716) when combined with atezolizumab (anti-PD-L1) versus atezolizumab alone1. However, there remains little consensus on the mechanism(s) of response with this combination2. Here we find that a high baseline of intratumoural macrophages and regulatory T cells is associated with better outcomes in patients treated with atezolizumab plus tiragolumab but not with atezolizumab alone. Serum sample analysis revealed that macrophage activation is associated with a clinical benefit in patients who received the combination treatment. In mouse tumour models, tiragolumab surrogate antibodies inflamed tumour-associated macrophages, monocytes and dendritic cells through Fcγ receptors (FcγR), in turn driving anti-tumour CD8+ T cells from an exhausted effector-like state to a more memory-like state. These results reveal a mechanism of action through which TIGIT checkpoint inhibitors can remodel immunosuppressive tumour microenvironments, and suggest that FcγR engagement is an important consideration in anti-TIGIT antibody development.

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

Access options

Buy this article

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

Fig. 1: Intratumoural myeloid and Treg cell content is associated with patient benefit after combination treatment with tiragolumab plus atezolizumab in the CITYSCAPE trial.
Fig. 2: Treatment with tiragolumab plus atezolizumab leads to increased serum myeloid proteins.
Fig. 3: Tiragolumab plus atezolizumab leads to T, NK and myeloid cell activation in PBMCs.
Fig. 4: Fc receptor engagement supports tiragolumab surrogate efficacy and ability to remodel the tumour microenvironment in mice.
Fig. 5: Flow cytometry analysis of anti-TIGIT antibody activity on tumour myeloid cells and lymphocytes.
Fig. 6: Macrophages enable modulation of CD8+ T cells by Fc-active anti-TIGIT antibodies in vivo and in vitro.

Similar content being viewed by others

Data availability

The sequencing data generated in this study will be deposited once anonymized. Up-to-date details on Roche’s Global Policy on the Sharing of Clinical Information and how to request access to related clinical study documents are available online (https://go.roche.com/data_sharing). Anonymized records for individual patients across more than one data source external to Roche cannot, and should not, be linked due to a potential increase in risk of patient re-identification. Source data of preclinical study data are provided with this paper, and source data of clinical study data will be deposited once anonymized. Source data are provided with this paper.

Code availability

All packages used in this study are publicly available. This study does not report original codes.

Change history

References

  1. Cho, B. C. et al. Tiragolumab plus atezolizumab versus placebo plus atezolizumab as a first-line treatment for PD-L1-selected non-small-cell lung cancer (CITYSCAPE): primary and follow-up analyses of a randomised, double-blind, phase 2 study. Lancet Oncol. 23, 781–792 (2022).

    Article  CAS  PubMed  Google Scholar 

  2. Dolgin, E. Antibody engineers seek optimal drug targeting TIGIT checkpoint. Nat. Biotechnol. 38, 1007–1009 (2020).

    Article  CAS  PubMed  Google Scholar 

  3. Sharma, P., Hu-Lieskovan, S., Wargo, J. A. & Ribas, A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 168, 707–723 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Gong, J., Le, T. Q., Massarelli, E., Hendifar, A. E. & Tuli, R. Radiation therapy and PD-1/PD-L1 blockade: the clinical development of an evolving anticancer combination. J. Immunother. Cancer 6, 46 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Wang, M., Herbst, R. S. & Boshoff, C. Toward personalized treatment approaches for non-small-cell lung cancer. Nat. Med. 27, 1345–1356 (2021).

    Article  CAS  PubMed  Google Scholar 

  6. Chiang, E. Y. & Mellman, I. TIGIT-CD226-PVR axis: advancing immune checkpoint blockade for cancer immunotherapy. J. Immunother. Cancer 10, e004711 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Herbst, R. S. et al. Atezolizumab for first-line treatment of PD-L1-selected patients with NSCLC. N. Engl. J. Med. 383, 1328–1339 (2020).

    Article  CAS  PubMed  Google Scholar 

  8. Felip, E. et al. Adjuvant atezolizumab after adjuvant chemotherapy in resected stage IB-IIIA non-small-cell lung cancer (IMpower010): a randomised, multicentre, open-label, phase 3 trial. Lancet 398, 1344–1357 (2021).

    Article  CAS  PubMed  Google Scholar 

  9. Johnston, R. J. et al. The immunoreceptor TIGIT regulates antitumor and antiviral CD8+ T cell effector function. Cancer Cell 26, 923–937 (2014).

    Article  CAS  PubMed  Google Scholar 

  10. Banta, K. L. et al. Mechanistic convergence of the TIGIT and PD-1 inhibitory pathways necessitates co-blockade to optimize anti-tumor CD8+ T cell responses. Immunity 55, 512–526 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Johnston, R. J., Lee, P. S., Strop, P. & Smyth, M. J. Cancer immunotherapy and the nectin family. Annu. Rev. Cancer Biol. 5, 203–219 (2021).

    Article  Google Scholar 

  12. Waight, J. D. et al. Selective FcɣR co-engagement on APCs modulates the activity of therapeutic antibodies targeting T cell antigens. Cancer Cell 33, 1033–1047 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Han, J. H. et al. Effective anti-tumor response by TIGIT blockade associated with FcγR engagement and myeloid cell activation. Front. Immunol. 11, 573405 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Preillon, J. et al. Restoration of T-cell effector function, depletion of Tregs, and direct killing of tumor cells: the multiple mechanisms of action of a-TIGIT antagonist antibodies. Mol. Cancer Ther. 20, 121–131 (2021).

    Article  CAS  PubMed  Google Scholar 

  15. Yu, X. et al. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat. Immunol. 10, 48–57 (2008).

    Article  PubMed  Google Scholar 

  16. Stanietsky, N. et al. The interaction of TIGIT with PVR and PVRL2 inhibits human NK cell cytotoxicity. Proc. Natl Acad. Sci. USA 106, 17858–17863 (2009).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. Patil, N. S. et al. Intratumoral plasma cells predict outcomes to PD-L1 blockade in non-small cell lung cancer. Cancer Cell 40, 289–300 (2022).

    Article  CAS  PubMed  Google Scholar 

  18. Rittmeyer, A. 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 389, 255–265 (2017).

    Article  PubMed  Google Scholar 

  19. Dige, A. et al. Soluble CD163, a specific macrophage activation marker, is decreased by anti-TNF-α antibody treatment in active inflammatory bowel disease. Scand. J. Immunol. 80, 417–423 (2014).

    Article  CAS  PubMed  Google Scholar 

  20. Davis, B. H. & Zarev, P. V. Human monocyte CD163 expression inversely correlates with soluble CD163 plasma levels. Cytometry B. Clin. Cytom. 63, 16–22 (2005).

    Article  PubMed  Google Scholar 

  21. Bendell, J. C. et al. Phase Ia/Ib dose-escalation study of the anti-TIGIT antibody tiragolumab as a single agent and in combination with atezolizumab in patients with advanced solid tumors. Cancer Res. 80, CT302 (2020).

  22. Liberzon, A. et al. The Molecular Signatures Database (MSigDB) Hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Herbst, R. S. et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 515, 563–567 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  24. Bar, N. et al. Differential effects of PD-L1 versus PD-1 blockade on myeloid inflammation in human cancer. JCI Insight 5, e129353 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Pello, O. M. et al. Role of c-MYC in alternative activation of human macrophages and tumor-associated macrophage biology. Blood 119, 411–421 (2012).

    Article  PubMed  Google Scholar 

  26. Lo, M. et al. Effector-attenuating substitutions that maintain antibody stability and reduce toxicity in mice. J. Biol. Chem. 292, 3900–3908 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Nimmerjahn, F. & Ravetch, J. V. Fcγ receptors: old friends and new family members. Immunity 24, 19–28 (2006).

    Article  CAS  PubMed  Google Scholar 

  28. Huang, A. Y. et al. The immunodominant major histocompatibility complex class I-restricted antigen of a murine colon tumor derives from an endogenous retroviral gene product. Proc. Natl Acad. Sci. USA 93, 9730–9735 (1996).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  29. DeNardo, D. G. et al. Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov. 1, 54–67 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Ries, C. H. et al. Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell 25, 846–859 (2014).

    Article  CAS  PubMed  Google Scholar 

  31. O’Brien, S. A. et al. Activity of tumor-associated macrophage depletion by CSF1R blockade is highly dependent on the tumor model and timing of treatment. Cancer Immunol. Immunother. 70, 2401–2410 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Cannarile, M. A. et al. Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J. Immunother. Cancer 5, 53 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Ribas, A. & Wolchok, J. D. Cancer immunotherapy using checkpoint blockade. Science 359, 1350–1355 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  34. Morad, G., Helmink, B. A., Sharma, P. & Wargo, J. A. Hallmarks of response, resistance, and toxicity to immune checkpoint blockade. Cell 184, 5309–5337 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Yofe, I. et al. Anti-CTLA-4 antibodies drive myeloid activation and reprogram the tumor microenvironment through FcγR engagement and type I interferon signaling. Nat. Cancer 3, 1336–1350 (2022).

    Article  CAS  PubMed  Google Scholar 

  36. Casanova-Acebes, M. et al. Tissue-resident macrophages provide a pro-tumorigenic niche to early NSCLC cells. Nature 595, 578–584 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  37. Nielsen, S. R. & Schmid, M. C. Macrophages as key drivers of cancer progression and metastasis. Mediators Inflamm. 2017, 9624760 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Haas, L. & Obenauf, A. C. Allies or enemies—the multifaceted role of myeloid cells in the tumor microenvironment. Front. Immunol. 10, 2746 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lindau, D., Gielen, P., Kroesen, M., Wesseling, P. & Adema, G. J. The immunosuppressive tumour network: myeloid-derived suppressor cells, regulatory T cells and natural killer T cells. Immunology 138, 105–115 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Zilionis, R. et al. Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity 50, 1317–1334 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Bournazos, S. et al. Signaling by antibodies: recent progress. Annu. Rev. Immunol. 35, 285–311 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Bruderer, R. et al. Analysis of 1508 plasma samples by capillary-flow data-independent acquisition profiles proteomics of weight loss and maintenance. Mol. Cell. Proteom. 18, 1242–1254 (2019).

    Article  CAS  Google Scholar 

  43. Callister, S. J. et al. Normalization approaches for removing systematic biases associated with mass spectrometry and label-free proteomics. J. Proteome Res. 5, 277–286 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Leek, J. T. & Storey, J. D. Capturing heterogeneity in gene expression studies by surrogate variable analysis. PLoS Genet. 3, 1724–1735 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Richie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  Google Scholar 

  46. Stoeckius, M. et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods 14, 865–868 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Oh, S. A. et al. PD-L1 expression by dendritic cells is a key regulator of T-cell immunity in cancer. Nat. Cancer 1, 681–691 (2020).

    Article  CAS  PubMed  Google Scholar 

  48. Mily, A. et al. Polarization of M1 and M2 human monocyte-derived cells and analysis with flow cytometry upon Mycobacterium tuberculosis infection. J. Vis. Exp. 163, e61807 (2020).

    Google Scholar 

  49. Wu, T. D., Reeder, J., Lawrence, M., Becker, G. & Brauer, M. J. GMAP and GSNAP for genomic sequence alignment: enhancements to speed, Accuracy, and Functionality. Methods Mol. Biol. 1418, 283–334 (2016).

    Article  PubMed  Google Scholar 

  50. Lawrence, M. et al. Software for computing and annotating genomic ranges. PLoS Comput. Biol. 9, e1003118 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Lambrechts, D. et al. Phenotype molding of stromal cells in the lung tumor microenvironment. Nat. Med. 24, 1277–1289 (2018).

    Article  CAS  PubMed  Google Scholar 

  52. Laboratory for Functional Epigenetics, gbiomed.kuleuven.be/english/cme/research/laboratories/54213024/scRNAseq-NSCLC (KU Leuven, 2023).

  53. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Cheng, S. et al. A pan-cancer single-cell transcriptional atlas of tumor infiltrating myeloid cells. Cell 184, 792–809 (2021).

    Article  CAS  PubMed  Google Scholar 

  56. Kim, N. et al. Single-cell RNA sequencing demonstrates the molecular and cellular reprogramming of metastatic lung adenocarcinoma. Nat. Commun. 11, 2285 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  57. Bagaev, A. et al. Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. Cancer Cell 39, 845–865 (2021).

    Article  CAS  PubMed  Google Scholar 

  58. Mariathasan, S. et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 554, 544–548 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  59. Sergushichev, A. A. An algorithm for fast preranked gene set enrichment analysis using cumulative statistic calculation. Preprint at bioRxiv https://doi.org/10.1101/060012 (2021).

Download references

Acknowledgements

We thank the patients who provided tumour samples for this study, as well as the investigators and staff involved in the CITYSCAPE study. Editorial assistance for the development of this Article, under the direction of the authors, was provided by A. Robertson and D. Christofi of Ashfield MedComms, an Inizio company, and funded by F. Hoffmann-La Roche. We also thank the staff at Biognosys for processing and generating the raw mass spectrometry data files for analysis; the staff at Abiosciences for generating mouse scRNA-seq raw data files; the staff at Immunai for generating and processing human PBMC scRNA-seq data; and C. Bais, T. Pham, R. Greathouse and A. Rapaport for reading the manuscript and providing technical assistance.

Author information

Authors and Affiliations

Authors

Contributions

X.G. and R. Hu are co-first authors; Y.C. and S.S. are co-second authors; and R.J.J. and N.S.P. are co-senior authors. N.S.P. and R.J.J. conceived the project. R. Hu, S.S., J.S., L.M., R. Hendricks, K.N., K.L.B., E.D., P.S.C., J.H. and S. Mittman performed experiments. N.S.P., R.J.J., X.G., R. Hu, Y.C., S.S., B.Y.N., L.M. and E.D analysed data. N.S.P., S.S., E.Y.C., L.M., A.D., S. Mariathasan, R.M., D.S.S., I.M. and R.J.J. guided data analysis. W.C., N.M. and P.D. coordinated clinical sample management and analysis. M.J., D.R.A., B.C.C., A.I., I.G.-B., E.F. and R.M. guided clinical trial and data management. N.S.P., R.J.J. and X.G. wrote the manuscript with input from all of the authors. All of the authors contributed to data interpretation, discussion of results and commented on the manuscript.

Corresponding authors

Correspondence to Robert J. Johnston or Namrata S. Patil.

Ethics declarations

Competing interests

X.G., R. Hu, Y.C., S.S., B.Y.N., J.S., L.M., R. Hendricks, K.N., K.L.B., E.D., A.D., P.S.C., J.H., S. Mittman, N.M., P.D., W.C., I.M., S. Mariathasan, D.S.S., R.M., E.Y.C., R.J.J. and N.S.P. are employees and stockholders of Roche/Genentech. M.J. declares research funding (paid to institution) from AbbVie, Acerta, Adaptimmune, Amgen, Apexigen, Arcus Biosciences, Array BioPharma, ArriVent BioPharma, Artios Pharma, AstraZeneca, Atreca, BeiGene, BerGenBio, BioAtla, Black Diamond, Boehringer Ingelheim, Bristol-Myers Squibb, Calithera Biosciences, Carisma Therapeutics, Checkpoint Therapeutics, City of Hope National Medical Center, Corvus Pharmaceuticals, Curis, CytomX, Daiichi Sankyo, Dracen Pharmaceuticals, Dynavax, Lilly, Eikon Therapeutics, Elicio Therapeutics, EMD Serono, EQRx, Erasca, Exelixis, Fate Therapeutics, Genentech/Roche, Genmab, Genocea Biosciences, GlaxoSmithKline, Gritstone Oncology, Guardant Health, Harpoon, Helsinn Healthcare SA, Hengrui Therapeutics, Hutchison MediPharma, IDEAYA Biosciences, IGM Biosciences, Immunitas Therapeutics, Immunocore, Incyte, Janssen, Jounce Therapeutics, Kadmon Pharmaceuticals, Kartos Therapeutics, LockBody Therapeutics, Loxo Oncology, Lycera, Memorial Sloan-Kettering, Merck, Merus, Mirati Therapeutics, Mythic Therapeutics, NeoImmune Tech, Neovia Oncology, Novartis, Numab Therapeutics, Nuvalent, OncoMed Pharmaceuticals, Palleon Pharmaceuticals, Pfizer, PMV Pharmaceuticals, Rain Therapeutics, RasCal Therapeutics, Regeneron Pharmaceuticals, Relay Therapeutics, Revolution Medicines, Ribon Therapeutics, Rubius Therapeutics, Sanofi, Seven and Eight Biopharmaceuticals/Birdie Biopharmaceuticals, Shattuck Labs, Silicon Therapeutics, Stem CentRx, Syndax Pharmaceuticals, Taiho Oncology, Takeda Pharmaceuticals, Tarveda, TCR2 Therapeutics, Tempest Therapeutics, Tizona Therapeutics, TMUNITY Therapeutics, Turning Point Therapeutics, University of Michigan, Vyriad, WindMIL Therapeutics and Y-mAbs Therapeutics; and consulting/advisory roles (paid to institution) for AbbVie, Amgen, Arcus Biosciences, Arrivent, Astellas, AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Calithera Biosciences, D3 Bio Limited, Daiichi Sankyo, Fate Therapeutics, Genentech/Roche, Genmab, Genocea Biosciences, Gilead Sciences, GlaxoSmithKline, Gritstone Oncology, Hookipa Biotech, Immunocore, Janssen, Jazz Pharmaceuticals, Lilly, Merck, Mirati Therapeutics, Molecular Axiom, Normunity, Novartis, Novocure, Pfizer, Pyramid Biosciences, Revolution Medicines, Sanofi-Aventis, SeaGen, Synthekine, Takeda Pharmaceuticals and VBL Therapeutics. D.R.A. reports personal payment/honoraria from Roche, AstraZeneca, Bristol-Myers Squibb, Merck Sharp & Dohme, Eli Lilly, Pfizer, and Novartis; and institutional support for attending meetings or travel from Roche, Bristol-Myers Squibb, Merck Sharp & Dohme and Novartis. BCC declares royalties from Champions Oncology, Crown Bioscience, Imagen and PearlRiver Bio; grants/research support/funding from MOGAM Institute, LG Chem, Oscotec, Interpark Bio Convergence Corp, GIInnovation, GI-Cell, Abion, Abbvie, AstraZeneca, Bayer, Blueprint Medicines, Boehringer Ingelheim, Champions Oncology, CJ Bioscience, CJ Blossom Park, Cyrus, Dizal Pharma, Genexine, Janssen, Lilly, MSD, Novartis, Nuvalent, Oncternal, Ono, Regeneron, Dong-A ST, Bridgebio Therapeutics, Yuhan, ImmuneOncia, Illumina, Kanaph Therapeutics, Therapex, JINTSbio, Hanmi, CHA Bundang Medical Center and Vertical Bio AG; consultancy roles for Abion, BeiGene, Novartis, AstraZeneca, Boehringer-Ingelheim, Roche, BMS, CJ, CureLogen, Cyrus Therapeutics, Ono, Onegene Biotechnology, Yuhan, Pfizer, Eli Lilly, GI-Cell, Guardant, HK Inno-N, Imnewrun Biosciences, Janssen, Takeda, MSD, Medpacto, Blueprint medicines, RandBio and Hanmi; employment from Yonsei University Health System; participation on an advisory board for KANAPH Therapeutic, Bridgebio Therapeutics, Cyrus Therapeutics, Guardant Health, Oscotec, J INTS Bio, Therapex, Gliead and Amgen; speaker roles for ASCO, AstraZeneca, Guardant, Roche, ESMO, IASLC, Korean Cancer Association, Korean Society of Medical Onoclogy, Korean Society of Thyroid-Head and Neck Surgery, Korean Cancer Study Group, Novartis, MSD, The Chinese Thoracic Oncology Society and Pfizer; stocks/shares in TheraCanVac, Gencurix, Bridgebio Therapeutics, KANAPH Therapeutic, Cyrus Therapeutics, Interpark Bio Convergence and J INTS BIO; founder for DAAN Biotherapeutics; and member of the board of directors for J INTS BIO. A.I. declares grants and/or consulting fees from BMS, MSD, Roche, Bayer and AstraZeneca. I.G.-B. declares clinical investigator, advisory board, speaker and director of scientific meeting roles for Roche/Genentech; and financial support from Roche/Genentech. E.F. declares advisory board/speaker roles for Abbvie, Amgen, Astra Zeneca, Bayer, Beigene, Boehringer Ingelheim, Bristol Myers Squibb, Daiichi Sankyo, Eli Lilly, F. Hoffmann-La Roche, Genentech, Gilead, Glaxo Smith Kline, Janssen, Medscape, Merck Serono, Merck Sharp & Dohme, Novartis, Peervoice, Peptomyc, Pfizer, Regeneron, Sanofi, Takeda, Touch Oncology and Turning Point Therapeutics; and independent member of the board for Grifols.

Peer review

Peer review information

Nature thanks Kristen Pauken and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Intratumoural myeloid and Treg cell content correlates with tiragolumab plus atezolizumab outcome but not placebo plus atezolizumab.

a, Forest plot comparing tiragolumab plus atezolizumab versus placebo plus atezolizumab in patients with tumours expressing high or low gene levels (cutoff by median expression) of CD274, TIGIT, CD226, and PVR in CITYSCAPE. Hazard ratio and 95% confidence interval were determined using univariate Cox model. The dots represent the hazard ratio and the horizontal bars the 95% confidence interval. b–e, Kaplan–Meier curves comparing PFS in patients with tumours enriched (solid lines) or not enriched (dashed lines) for TAMs (b), Tregs (c), CD16-high monocytes (d), and CD8 + T effector cells (T-eff) (e). Enrichment or not was determined by the median cell type signature score cutoffs. f, g, Kaplan–Meier curves comparing the PFS (f) and OS (g) in PD-L1-positive patients from the phase 3 NSCLC OAK study who received atezolizumab monotherapy and had tumours enriched for TAMs. h, i, Kaplan–Meier curves comparing the PFS (h) and OS (i) in PD-L1-positive patients from the phase 3 NSCLC OAK study who received atezolizumab monotherapy and had tumours enriched for Tregs. f-i, Hazard ratio and 95% confidence interval were determined using univariate Cox model, and P values were estimated using the log-rank test.

Extended Data Fig. 2 Correlation of bulk RNA-seq-based cell type signature scores with multiplex immunofluorescence.

a, b, Correlation of TAM signature with CD68+ cells by mIF (a) and Treg signature with FoxP3+ cells by mIF (b). Two-tailed Pearson correlation; n = 27. mIF, multiplex immunofluorescence.

Extended Data Fig. 3 The proportion of proliferation cells and major cell types in PBMC.

a, Scatter plot showing the S and G2M cell cycle phase scores, coloured by cells in proliferating (red) or non-proliferating states (black). b, Bar plot showing the proportion of proliferating cells in each major cell type. c, Box plots showing the proportion of proliferating cells in CD4_non_naive, CD8_non_naive, and NK cells, across each timepoint. d, Box plots comparing the proportions of each cell type at on-treatment (C1D15, C2D1, and C4D1) versus baseline (C1D1). e, Box plots comparing the proportions of each cell type between responders and non-responders at baseline (C1D1) and on-treatment (C1D15, C2D1, and C4D1). c-e, Boxplot center line, median; box, interquartile range (IQR; the range between the 25th and 75th percentile); whiskers, 1.58 × IQR. c,d, Median values per time point are connected by solid black lines; samples from the same patient at different time points are connected by grey lines. c-d, P values shown were calculated by two-tailed paired Student’s t-test and BH-adjusted. e, Nominal P values derived from two-tailed unpaired Student t-test are shown and red asterisk represents significance levels where * P < 0.05. c-e, n = 16 patients.

Source Data

Extended Data Fig. 4 Efficient tumour rejection by anti-PD-L1 and anti-TIGIT mAbs treatment depends on functional Fc-FcɣR interaction axis.

a, Plots depicting tumour volumes in each mouse over time; data are representative of one independent experiment. Wildtype BALB/c mice were implanted with CT26 tumours and then treated as described in the method. b, Plots depicting tumour volumes in each mouse over time; data are representative of one independent experiment. Wildtype (top) and FcɣR knockout (bottom) BALB/c mice were implanted with CT26 tumours and then treated as described in the method.

Source Data

Extended Data Fig. 5 anti-TIGIT treatment modulation of tumour infiltrating immune cells and peripheral blood monocytes depends on the Fc region.

a, UMAP of single cells from tumour infiltrating T and NK cells (top, n = 21,407) and myeloid cells (bottom, n = 5,352) coloured by cell types. b, Bubble plots showing marker gene expression for T and NK cells (left) and myeloid cells (right) as shown in (a).c-e, Heatmaps showing the expression of selected genes across different treatments in tumour macrophages and monocytes combined (c), tumour CD8 + T cells combined (d), and tumour CD4+ Tregs (e). f, UMAP of single cells from the peripheral blood (n = 26,174) coloured by cell types. g, Bubble plots showing the marker gene expression of cell types as in (f). h, Heatmap displaying the scaled gene expression of marker genes distinguishing classical, non-classical, and intermediate monocytes, and the expression patterns of FcɣR. i, Heatmap showing the scaled gene expression of MHC and interferon response in non-classical monocytes across different treatments. a-i, Single cell RNA-seq was performed on intratumoural (a-e) and peripheral (f-i) CD45+ cells isolated at day 3 after treatment, and data are from one independent experiment with n = 5 mice in each group.

Source Data

Extended Data Fig. 6 Annotation of single cells collected from mouse tumours.

Single cell RNA-seq was performed on intratumoural CD45+ cells isolated from tumours at day 3 after treatment, and data are from one independent experiment with n = 5 mice in each group. This is related to Fig. 4b–d. a, UMAP of tumour-infiltrating lymphocytes (top, n = 35,358) and myeloid (bottom, n = 4,261) cells coloured by cell types. b, Bubble plots showing marker gene expression for T and NK cells (left) and myeloid cells (right) as shown in (a).

Source Data

Extended Data Fig. 7 The modulation effects of anti-PD-L1 + anti-TIGIT on peripheral blood monocytes depends on the anti-TIGIT mAb Fc region.

a, UMAP of single cells from the peripheral blood cells (n = 55,368) coloured by cell types. b, Bubble plot showing the marker gene expression of cell types as in (a). c, Heatmap displaying the scaled gene expression of marker genes distinguishing classical, non-classical, and intermediate monocytes, and the expression patterns of FcɣR. d, Box plots comparing cell proportions of different treatments versus IgG2a isotype control (B1). B2, aPD-L1; B3, aTIGIT-IgG2b; B4, aTIGIT-IgG2a; B5, aPD-L1 + aTIGIT-IgG2b; B6, aPD-L1 + aTIGIT-IgG2a. Boxplot center line, median; box, interquartile range (IQR; the range between the 25th and 75th percentile); whiskers, 1.58 × IQR. Normal P values by two-tailed unpaired Student’s t-test are shown in grey colour; adjusted P values by Dunnett’s multiple comparison were shown in black colour. e, f, Volcano plots showing the gene expression of anti-PD-L1 + anti-TIGIT IgG2a versus anti-PD-L1 (e), and anti-PD-L1 + anti-TIGIT-IgG2b versus anti-PD-L1 (f) in peripheral blood classical (left), intermediate (middle), and non-classical (right) monocytes. P values were calculated by two-tailed Wilcoxon rank-sum test. a-f, Single cell RNA-seq was performed on peripheral CD45+ cells isolated at day 3 after treatment, and data are from one independent experiment with n = 5 mice in each group.

Source Data

Extended Data Fig. 8 Flow cytometry analysis of anti-TIGIT activity in tumour myeloid cells of E0771 model, and T cells of CT26 model.

a, Mean fluorescence intensity (MFI) of cell surface MHC-II on tumour-infiltrating dendritic cells (DC, left), macrophages (middle), and monocytes (right), normalized to their respective median MFI value following control treatment. Far right, histogram of representative surface MHC-II expression on tumour monocytes following various treatments. E0771-bearing C57BL/6 J mice were treated as indicated and data were collected at day 7 after treatment. Data are a composite of two independent experiments with n = 4 mice in each group; shown are mean +/− SEM with one-way ANOVA with Dunnett’s multiple comparisons, with the Control IgG group designated as the control group. b, Frequencies of tumour-infiltrating FoxP3- non-Treg CD4 + T cells (left), FoxP3+ Treg CD4 + T cells (middle), and CD8 + T cells (right) out of total CD45+ cells. c, Ratio of tumour CD8 + T cells to FoxP3+ Treg CD4 + T cells. d, e, Additional data related to Fig. 5e,f. Frequencies of TCF1 + TIM3+ memory-like T cells (d) and TOX+ terminally differentiated effector T cells (e) in CT26-tumour bearing mice treated with control and anti-PD-L1 plus anti-TIGIT mIgG2a-LALAPG or mIgG2a antibodies. b-e, Intratumoural CD45+ cells were analysed by flow cytometry at day 3 after treatment (b, c) and gp70 tetramer positive T cells at day 7 after treatment (d, e). Data are representative of one (b, c) or two (d, e) independent experiments with n = 5 mice in each group. b-e, Data in the dot plots are mean +/− SEM with one-way ANOVA with Dunnett’s multiple comparisons, with the anti-PD-L1 monotherapy group designated as the control group.

Source Data

Extended Data Fig. 9 Tumour infiltrating leukocyte FACS and scRNA-seq analysis following treatment with anti-PD-L1, anti-TIGIT, and anti-CSF-1R.

a, Percentage of tumour macrophages (left) and representative FACS plots of tumour CD11b+ cell expression of F4/80 and CD86 following treatment (right). Data were collected at day 7 after treatment, and are representative of two independent experiments with n = 5 mice in each group. Left, data are mean +/ − SEM with one-way ANOVA with Tukey’s multiple comparisons. b, Growth of CT26 tumours in syngeneic BALB/c mice treated with anti-gp120 (left), anti-PD-L1 + anti-TIGIT IgG2a (middle), and anti-PD-L1 + anti-TIGIT mIgG2a + anti-CSF-1R (right). Data are representative of two experiments with n = 10 mice in each group. c, UMAP of tumour-infiltrating lymphocytes (top, n = 21, 575) and myeloid (bottom, n = 3, 734) cells coloured by cell types. d, Bubble plots showing marker gene expression for T and NK cells (left) and myeloid cells (right) as shown in (c). e, Volcano plots showing the gene expression of anti-PD-L1 + anti-TIGIT IgG2a versus control IgG2a (left), anti-PD-L1 + anti-TIGIT IgG2a + anti-CSF-1R versus control IgG2a (middle), and anti-PD-L1 + anti-TIGIT IgG2a + anti-CSF-1R versus anti-PD-L1 + anti-TIGIT IgG2a (right) in tumour macrophage and monocytes combined. f, g, Volcano plots showing the gene expression of anti-PD-L1 + anti-TIGIT IgG2a + anti-CSF-1R versus anti-PD-L1 + anti-TIGIT IgG2a in tumour CD8 + T cells combined (f) and CD4 Tregs (g). c-g, Single cell RNA-seq was performed on intratumoural CD45+ cells isolated from tumours at day 3 after treatment, and data are from one independent experiment with n = 5 mice in each group. In volcano plots, the broken y-axis was used to make the y-axis range comparable and for better comparison between treatments; P values were calculated by two-tailed Wilcoxon rank-sum test.

Source Data

Extended Data Fig. 10 Graphic illustration showing the design of the current study.

Top, To understand the mechanism(s) of response with tiragolumab in combination with atezolizumab, we leveraged samples collected from CITYSCAPE (left; NSCLC, Ph2) including tumour pretreatment samples for bulk RNA-seq and multiplex immunofluorescence (mIF), and pretreatment and on-treatment serum samples for Mass Spec, GO30103 (middle; NSCLC, Ph1b) including pretreatment and on-treatment peripheral blood mononuclear cells (PMBC) for single cell RNA-seq, and preclinical models (right). Bottom, Anti-TIGIT antibody, in a Fc dependent manner, remodels immunosuppressive tumour microenvironments by leveraging myeloid cells and Tregs, which was further enhanced with the addition of anti-PD-(L)1 antibody. Created with BioRender.com.

Supplementary information

Supplementary Figures

Supplementary Figs. 1–7.

Reporting Summary

Supplementary Table 1

Baseline characteristics in CITYSCAPE (ITT population and the BEP).

Supplementary Table 2

Population demographics in patients whose tumors have high vs low cell types. P values were derived using Fisher’s exact test.

Source data

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

Guan, X., Hu, R., Choi, Y. et al. Anti-TIGIT antibody improves PD-L1 blockade through myeloid and Treg cells. Nature 627, 646–655 (2024). https://doi.org/10.1038/s41586-024-07121-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-024-07121-9

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research