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:

The GPCR–Gαs–PKA signaling axis promotes T cell dysfunction and cancer immunotherapy failure

Abstract

Immune checkpoint blockade (ICB) targeting PD-1 and CTLA-4 has revolutionized cancer treatment. However, many cancers do not respond to ICB, prompting the search for additional strategies to achieve durable responses. G-protein-coupled receptors (GPCRs) are the most intensively studied drug targets but are underexplored in immuno-oncology. Here, we cross-integrated large singe-cell RNA-sequencing datasets from CD8+ T cells covering 19 distinct cancer types and identified an enrichment of Gαs-coupled GPCRs on exhausted CD8+ T cells. These include EP2, EP4, A2AR, β1AR and β2AR, all of which promote T cell dysfunction. We also developed transgenic mice expressing a chemogenetic CD8-restricted Gαs–DREADD to activate CD8-restricted Gαs signaling and show that a Gαs–PKA signaling axis promotes CD8+ T cell dysfunction and immunotherapy failure. These data indicate that Gαs–GPCRs are druggable immune checkpoints that might be targeted to enhance the response to ICB immunotherapies.

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: Gαs–GPCR correlation with T cell dysfunction and terminally exhausted T cells.
Fig. 2: Gαs coupling augments an exhaustion-like dysfunctional state in CD8+ T cells.
Fig. 3: Mechanisms of immune suppression in CD8+ T cells uncovered by Gαs–DREADD.
Fig. 4: CD8-restricted Gαs stimulation leads to immunotherapy failure.
Fig. 5: Gαs–GPCRs in individuals with cancer correlate with decreased survival and ICB response.
Fig. 6: The Gαs signaling axis as an immune checkpoint in cancer.

Similar content being viewed by others

Data availability

Gene Expression Omnibus accession numbers for the human datasets used in this study include GSE116256, GSE114727, GSE110686, GSE14018, GSE123813, GSE22898, GSE123139, GSE115978, GSE146771, GSE134520, GSE140228, GSE125449, GSE117570, GSE127465, GSE99254, GSE123813, GSE156728, GSE120575, GSE84820, GSE123235, GSE122969, GSE88987 and GSE141299. European Genome–Phenome Archive identifiers for the human datasets used in this study include EGAS00001002171, EGAS00001002486, EGAS00001002325 and EGAS00001002553. Additional information regarding these datasets can be found in Supplementary Table 1. The remaining data are available within the article and supplementary information. Source data are provided with this paper.

References

  1. Okazaki, T. & Honjo, T. PD-1 and PD-1 ligands: from discovery to clinical application. Int. Immunol. 19, 813–824 (2007).

    CAS  PubMed  Google Scholar 

  2. Leach, D. R., Krummel, M. F. & Allison, J. P. Enhancement of antitumor immunity by CTLA-4 blockade. Science 271, 1734–1736 (1996).

    CAS  PubMed  Google Scholar 

  3. Mellman, I., Coukos, G. & Dranoff, G. Cancer immunotherapy comes of age. Nature 480, 480–489 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Larkin, J. et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 373, 23–34 (2015).

    PubMed  PubMed Central  Google Scholar 

  5. Haslam, A. & Prasad, V. Estimation of the percentage of US patients with cancer who are eligible for and respond to checkpoint inhibitor immunotherapy drugs. JAMA Netw. Open 2, e192535 (2019).

    PubMed  PubMed Central  Google Scholar 

  6. Hauser, A. S., Attwood, M. M., Rask-Andersen, M., Schioth, H. B. & Gloriam, D. E. Trends in GPCR drug discovery: new agents, targets and indications. Nat. Rev. Drug Discov. 16, 829–842 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. O’Hayre, M., Degese, M. S. & Gutkind, J. S. Novel insights into G protein and G protein-coupled receptor signaling in cancer. Curr. Opin. Cell Biol. 27, 126–135 (2014).

    PubMed  PubMed Central  Google Scholar 

  8. Wu, V. et al. Illuminating the Onco-GPCRome: novel G protein-coupled receptor-driven oncocrine networks and targets for cancer immunotherapy. J. Biol. Chem. 294, 11062–11086 (2019).

  9. Pierce, K. L., Premont, R. T. & Lefkowitz, R. J. Seven-transmembrane receptors. Nat. Rev. Mol. Cell Biol. 3, 639–650 (2002).

    CAS  PubMed  Google Scholar 

  10. Tokunaga, R. et al. CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation—a target for novel cancer therapy. Cancer Treat. Rev. 63, 40–47 (2018).

    CAS  PubMed  Google Scholar 

  11. Ayers, M. et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Invest. 127, 2930–2940 (2017).

    PubMed  PubMed Central  Google Scholar 

  12. Yao, C. et al. Prostaglandin E2–EP4 signaling promotes immune inflammation through TH1 cell differentiation and TH17 cell expansion. Nat. Med. 15, 633–640 (2009).

    CAS  PubMed  Google Scholar 

  13. Vigano, S. et al. Targeting adenosine in cancer immunotherapy to enhance T-cell function. Front. Immunol. 10, 925 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Valero, C. et al. Response rates to anti-PD-1 immunotherapy in microsatellite-stable solid tumors with 10 or more mutations per megabase. JAMA Oncol. 7, 739–743 (2021).

    PubMed  Google Scholar 

  15. Andreatta, M. et al. Interpretation of T cell states from single-cell transcriptomics data using reference atlases. Nat. Commun. 12, 2965 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Nieto, P. et al. A single-cell tumor immune atlas for precision oncology. Genome Res. 31, 1913–1926 (2021).

    PubMed  PubMed Central  Google Scholar 

  17. Zheng, L. et al. Pan-cancer single-cell landscape of tumor-infiltrating T cells. Science 374, abe6474 (2021).

    PubMed  Google Scholar 

  18. Li, H. et al. Dysfunctional CD8 T cells form a proliferative, dynamically regulated compartment within human melanoma. Cell 176, 775–789 (2019).

    CAS  PubMed  Google Scholar 

  19. Zander, R. et al. CD4+ T cell help is required for the formation of a cytolytic CD8+ T cell subset that protects against chronic infection and cancer. Immunity 51, 1028–1042 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Wherry, E. J. et al. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 27, 670–684 (2007).

    CAS  PubMed  Google Scholar 

  21. Wherry, E. J. & Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 15, 486–499 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Inoue, A. et al. Illuminating G-protein-coupling selectivity of GPCRs. Cell 177, 1933–1947 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Liu, C. et al. Neuropilin-1 is a T cell memory checkpoint limiting long-term antitumor immunity. Nat. Immunol. 21, 1010–1021 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Andrews, L. P. et al. A Cre-driven allele-conditioning line to interrogate CD4. Immunity 54, 2209–2217 (2021).

    CAS  PubMed  Google Scholar 

  25. Iglesias-Bartolome, R. et al. Inactivation of a Gαs–PKA tumour suppressor pathway in skin stem cells initiates basal-cell carcinogenesis. Nat. Cell Biol. 17, 793–803 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Roth, B. L. DREADDs for neuroscientists. Neuron 89, 683–694 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Nagai, Y. et al. Deschloroclozapine, a potent and selective chemogenetic actuator enables rapid neuronal and behavioral modulations in mice and monkeys. Nat. Neurosci. 23, 1157–1167 (2020).

    CAS  PubMed  Google Scholar 

  28. Zhang, Y. et al. MKP-1 is necessary for T cell activation and function. J. Biol. Chem. 284, 30815–30824 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Lang, R. & Raffi, F. A. M. Dual-specificity phosphatases in immunity and infection: an update. Int. J. Mol. Sci. 20, 2710 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Seo, H. et al. TOX and TOX2 transcription factors cooperate with NR4A transcription factors to impose CD8. Proc. Natl Acad. Sci. USA 116, 12410–12415 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Thommen, D. S. & Schumacher, T. N. T cell dysfunction in cancer. Cancer Cell 33, 547–562 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Wang, Z. et al. Syngeneic animal models of tobacco-associated oral cancer reveal the activity of in situ anti-CTLA-4. Nat. Commun. 10, 5546 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Sade-Feldman, M. et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Samstein, R. M. et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat. Genet. 51, 202–206 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Sinha, N. et al. Immune Determinants of the Association between Tumor Mutational Burden and Immunotherapy Response across Cancer Types. Cancer Res. 82, 2076–2083 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Miller, B. C. et al. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol. 20, 326–336 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Zelenay, S. et al. Cyclooxygenase-dependent tumor growth through evasion of immunity. Cell 162, 1257–1270 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Bottcher, J. P. et al. NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control. Cell 172, 1022–1037 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Pelly, V. S. et al. Anti-inflammatory drugs remodel the tumor immune environment to enhance immune checkpoint blockade efficacy. Cancer Discov. 11, 2602–2619 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Daher, C. et al. Blockade of β-adrenergic receptors improves CD8+ T-cell priming and cancer vaccine efficacy. Cancer Immunol. Res. 7, 1849–1863 (2019).

    PubMed  Google Scholar 

  41. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    CAS  PubMed  Google Scholar 

  42. Saddawi-Konefka, R. et al. Lymphatic-preserving treatment sequencing with immune checkpoint inhibition unleashes cDC1-dependent antitumor immunity in HNSCC. Nat. Commun. 13, 4298 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Belteki, G. et al. Conditional and inducible transgene expression in mice through the combinatorial use of Cre-mediated recombination and tetracycline induction. Nucleic Acids Res. 33, e51 (2005).

    PubMed  PubMed Central  Google Scholar 

  44. Akhmedov, D. et al. Gs–DREADD knock-in mice for tissue-specific, temporal stimulation of cyclic AMP signaling. Mol. Cell. Biol. 37, e00584-16 (2017).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    PubMed  PubMed Central  Google Scholar 

  48. Singh, G., Inoue, A., Gutkind, J. S., Russell, R. B. & Raimondi, F. PRECOG: predicting coupling probabilities of G-protein coupled receptors. Nucleic Acids Res. 47, W395–W401 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Yu, G., Wang, L. G., Han, Y. & He, Q. Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Valero, C. et al. Pretreatment neutrophil-to-lymphocyte ratio and mutational burden as biomarkers of tumor response to immune checkpoint inhibitors. Nat. Commun. 12, 729 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This project was supported by grants from the National Cancer Institute (R01CA247551 and U54CA209891) and National Institute of Dental and Craniofacial Research (NIH/NIDCR, R01DE026870 and U01DE028227). V.H.W. was supported by an NRSA Training Award (NIH/NCI 1F31CA250488-01). R.S.-K. was supported by an NRSA Training Award (NIH/NIDCR F32DE029990-01). B.S.Y. was supported by the Pharmacological Sciences Training Program (5T32GM007752-40) and an NRSA Training Award (NIH/NIDCR1F31DE031961-01). J.P.M. was supported by grants from the National Cancer Institute (U24CA248457 and U24CA220341). A.T.W. was supported by grants from the National Cancer Institute (F31CA257344 and U54CA209891) and the National Library of Medicine (T15LM011271). R.B. was supported by grants from the National Institutes of Health NIDDK (R01-DK092590) and NIAMS (R01-AR-072368). We acknowledge A. Sharabi (University of California, San Diego) for gifting the MC38-OVA cell line, D. Vignali (University of Pittsburg) for gifting the E8iCreErt2 mouse model, R. Iglesias-Bartolome (National Cancer Institute) for gifting the Gnas-exon 1fl/fl and the Tet-GFP-PKI mouse models and R. Berdeaux (The University of Texas) for gifting the ROSA26LSLGsDREADD mice. We acknowledge A. Goldrath for expertise and guidance in the study design and direction of the project.

Author information

Authors and Affiliations

Authors

Contributions

V.H.W. and B.S.Y. designed and conceived the studies, conducted most experiments described in this study and interpreted the data. F.F., Z.W. and R.S.-K. performed experiments described in the study. A.T.W., M.J.S., M.S.P., L.M.C., J.C., S.S., M.M., F.R., H.C., E.R. and J.P.M. all contributed to the bioinformatics analysis of this project. V.H.W., B.S.Y., F.F., R.S.-K., T.S.H., D.A.A.V., R.I.-B., R.B. and J.S.G. contributed to the study design and writing of the manuscript. J.S.G. provided oversight and direction of the entire project and study design, provided financial support for the study, interpreted the data and wrote the manuscript.

Corresponding author

Correspondence to J. Silvio Gutkind.

Ethics declarations

Competing interests

J.S.G. reports consulting fees from Domain Pharmaceuticals, Pangea Therapeutics and io9 and is founder of Kadima Pharmaceuticals, unrelated to the current study. R.B. is an employee and shareholder of CellChorus, Inc. J.C. is an employee and shareholder of Pfizer, Inc. V.H.W. is an employee and shareholder of Septerna, Inc. The other authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks William Murphy, Carla Rothlin, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: N. Bernard, in collaboration with the Nature Immunology 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 Patient and cancer information for integration analysis of CD8 T cells.

a, Information about patients, cancer type, and dataset used in the singe-cell RNA-seq integration. b, Visualization of 217,953 CD8 T cells after integration from 30 singe-cell RNA-seq datasets. c, Statistical comparison of calculated dysfunction score from tumor-infiltrating populations of CD8s characterized from Fig. 1b. Each dot represents one cell from groups listed in Supplementary Table 1a. Naïve: lower bound=0.147, middle bound=0.229, upper bound=0.337, 25th percentile=0.228, 75th percentile=0.231. Proliferating: lower bound=0.372, middle bound=0.564, upper bound=0.790, 25th percentile=0.556, 75th percentile=0.571. Cytotoxic: lower bound=0.184, middle bound=0.290, upper bound=0.447, 25th percentile=0.288, 75th percentile=0.292. Effector Memory: lower bound=0.209, middle bound=0.304, upper bound=0.431, 25th percentile=0.303, 75th percentile=0.305. Exhausted: lower bound=0.361, middle bound=0.555, upper bound=0.800, 25th percentile=0.551, 75th percentile=0.559.

Source data

Extended Data Fig. 2 Effect of targeting the Gαs/PKA signaling pathway in CD8 T cells.

a, Upregulation of inhibitory receptors and decrease of IFNγ and TNF in chronically versus acutely simulated CD8 T cells. The average relative expression and s.e.m. are shown (n = 6 biologically independent samples). b, Significant decrease of IFNγ or TNF with Gαs agonists in chronically stimulated CD8 T cells. The average relative expression and s.e.m. are shown (n = 6 biologically independent samples). c, Significant decrease of Ki-67 and viability with Gαs agonists in chronically stimulated CD8 T cells. The average relative expression and s.e.m. are shown (n = 6 biologically independent samples). d, Representative flow cytometry plots showing expression of Tim-3 and PD-1 in chronically stimulated CD8 T cells after treatment with 1 µM PGE2 (P), 5 µM Dobutamine (D), or 5 µM CGS-21860 (C). e, Effect of CXCL10 on PGE2-mediated decrease in IFNγ and TNFα The average frequency and s.e.m. are shown (n = 3 per group). Statistical significance was determined by two-way ANOVA. Unless indicated otherwise, statistical significance was determined by two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 3 Development of a CD8-restricted PKI transgenic mouse model.

a, Scheme illustrating the generation of CD8-PKI mice. b, Genotyping information for CD8-PKI mice. c, Confirmation of PKI expression in CD4 or CD8 T cells isolated from splenocytes of CD8-PKI mice and littermate controls after induction by doxycycline and tamoxifen. The average relative expression and s.e.m. are shown (n = 6 mice per group). d, Quantification of IFNγ and TNF inhibition by PGE2 in chronically stimulated CD8 T cells from CD8-PKI mice. The average relative expression and s.e.m. are shown (n = 3 biologically independent samples). Statistical significance was determined by two-way ANOVA.

Source data

Extended Data Fig. 4 Development of a CD8-restricted Gαs-DREADD transgenic mouse model.

a, Genotyping confirmation for CD8-GsD mice. Primers detecting the Gαs-DREADD, ROSA26, and E8i-Cre were used to confirm recombination by the Cre-recombinase. Information about primers and genotyping is listed in Supplementary Table 4. b, Effect of DCZ on circulating CD8, CD4, NK cells, and CD11b myeloid cells in the peripheral blood of CD8-GsD mice treated with tamoxifen and 5 doses of DCZ (n = 5 mice for -DCZ; n = 6 mice for +DCZ). c, Effect of DCZ on non-tamoxifen-treated CD8-GsD mice. Quantification of IFNγ and TNF and PD-1 and Tim-3 in non-tamoxifen-treated CD8 T cells treated with or without DCZ. The average frequency and s.e.m. are shown (n = 3 biologically independent samples). Statistical significance was determined by two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 5 Effect of tamoxifen and DCZ on tumor growth.

Tumor growth curve of CD8-GsD littermate control mice implanted with 4MOSC1 tumors treated with or without tamoxifen or DCZ. Mice were given 3 doses of tamoxifen, and 5 × 105 4MOSC1 cells were implanted into the tongue. Where indicated, 0.01 mg/kg DCZ was administered daily starting one day after tumor implantation.

Source data

Extended Data Fig. 6 Anti-CTLA-4 response in CD8-GsD mice bearing 4MOSC1 tumors.

Tumor growth curve (left panel) and survival plot (right panel) of CD8-GsD mice implanted with 4MOSC1 tumors treated with anti-CTLA-4 with or without DCZ (n = 7 mice per group). Mice were given three doses of tamoxifen before orthotopic tumor implantation and treated with checkpoint inhibitors and DCZ as previously described. Statistical significance was determined by two-way ANOVA. Statistical significance of survival data was calculated by the log-rank test.

Source data

Extended Data Fig. 7 Effect of Gnas deletion CD8 T cells in mice bearing 4MOSC1 tumors.

a, Tumor growth curve (left panel) and quantification of endpoint tumor volume (right panel) of CD8-Gnas+/+ (n = 14 mice) and CD8-Gnas−/− mice ( = 12 mice) implanted with 4MOSC1 tumors. Mice were given 3 doses of tamoxifen prior to orthotopic tumor implantation. The average tumor volume and s.e.m. are shown. Statistical significance was determined by two-way ANOVA. b, Quantification of PD-1+TIGIT+ CD8 T cells in 4MOSC1 tumors and draining lymph nodes at endpoint. The average frequency and s.e.m. are shown (n = 5 mice per group). Statistical significance was determined by two-tailed unpaired Student’s t-test. c, Frequency of CD8+ T cells in 4MOSC1 tumors in CD8-Gnas KO mice versus littermate controls. The average frequency and s.e.m. are shown (n = 5 mice per group). Statistical significance was determined by two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 8 Gating strategy for chronic stimulation and in vivo experiments.

a, For chronic stimulation experiments, lymphocytes were gated from forward scatter area (FSC-A) and side scatter area (SSC-A). Single cells were distinguished from doublet cells in forward scatter height (FSC-H) and forward scatter width (FSC-W), and then side scatter height (SSC-H) and side scatter width (SSC-W). Live CD8 cells were then gated. b, For in vivo experiments, lymphocytes were gated from forward scatter area (FSC-A) and side scatter area (SSC-A). Single cells were distinguished from doublet cells in forward scatter height (FSC-H) and forward scatter width (FSC-W), and then side scatter height (SSC-H) and side scatter width (SSC-W). Live CD45 cells were then gated. T cells were distinguished by NK1.1 negative, CD19 negative, and CD3 positive. CD8 T cells were then gated as CD4 negative.

Supplementary information

Reporting Summary

Supplementary Table

Supplementary Tables 1–6.

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

Wu, V.H., Yung, B.S., Faraji, F. et al. The GPCR–Gαs–PKA signaling axis promotes T cell dysfunction and cancer immunotherapy failure. Nat Immunol 24, 1318–1330 (2023). https://doi.org/10.1038/s41590-023-01529-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-023-01529-7

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