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.

  • Letter
  • Published:

An adaptive, biomarker-directed platform study of durvalumab in combination with targeted therapies in advanced urothelial cancer

Abstract

Durvalumab is a programmed death-ligand 1 (PD-L1) inhibitor with clinical activity in advanced urothelial cancer (AUC)1. AUC is characterized by several recurrent targetable genomic alterations2,3,4,5. This study (NCT02546661, BISCAY) combined durvalumab with relevant targeted therapies in biomarker-selected chemotherapy-refractory AUC populations including: (1) fibroblast growth factor receptor (FGFR) inhibitors in tumors with FGFR DNA alterations (FGFRm); (2) pharmacological inhibitor of the enzyme poly-ADP ribose polymerase (PARP) in tumors with and without DNA homologous recombination repair deficiency (HRRm); and (3) TORC1/2 inhibitors in tumors with DNA alteration to the mTOR/PI3K pathway3,4,5.This trial adopted a new, biomarker-driven, multiarm adaptive design. Safety, efficacy and relevant biomarkers were evaluated. Overall, 391 patients were screened of whom 135 were allocated to one of six study arms. Response rates (RRs) ranged 9–36% across the study arms, which did not meet efficacy criteria for further development. Overall survival (OS) and progression-free survival (PFS) were similar in the combination arms and durvalumab monotherapy arm. Biomarker analysis showed a correlation between circulating plasma-based DNA (ctDNA) and tissue for FGFRm. Sequential circulating tumor DNA analysis showed that changes to FGFRm correlated with clinical outcome. Our data support the clinical activity of FGFR inhibition and durvalumab monotherapy but do not show increased activity for any of the combinations. These findings question the targeted/immune therapy approach in AUC.

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: Biomarker landscape of enrolled patients in tumor and plasma.
Fig. 2: Efficacy data for different arms of the BISCAY study.
Fig. 3: Longitudinal mutation tracking on treatment.
Fig. 4: Correlation of FGFR mutant allele fraction ctDNA and response to therapy.

Similar content being viewed by others

Data availability

The authors declare that access to the clinical and biomarker data analyzed here may be obtained in accordance with AstraZeneca’s data-sharing policy as part of an external collaborative request (https://astrazenecagroup-dt.pharmacm.com//DT/Home/Index/) or an external data access request (https://vivli.org/ourmember/astrazeneca/)

References

  1. Powles, T. et al. Efficacy and safety of durvalumab in locally advanced or metastatic urothelial carcinoma: updated results from a phase 1/2 open-label study. JAMA Oncol. 3, e172411 (2017).

    Article  Google Scholar 

  2. Grivas, P. & Yu, E. Y. Role of targeted therapies in management of metastatic urothelial cancer in the era of immunotherapy. Curr. Treat. Options Oncol. 20, 67 (2019).

    Article  Google Scholar 

  3. Robertson, A. G. et al. Comprehensive molecular characterization of muscle-invasive bladder cancer. Cell 171, 540–556 (2017).

    Article  CAS  Google Scholar 

  4. Garcia, J. A. & Danielpour, D. Mammalian target of rapamycin inhibition as a therapeutic strategy in the management of urologic malignancies. Mol. Cancer Ther. 7, 1347–1354 (2008).

    Article  CAS  Google Scholar 

  5. Teo, M. Y. et al. DNA damage response and repair gene alterations are associated with improved survival in patients with platinum-treated advanced urothelial carcinoma. Clin. Cancer Res. 23, 3610–3618 (2017).

    Article  CAS  Google Scholar 

  6. von der Maase, H. et al. Gemcitabine and cisplatin versus methotrexate, vinblastine, doxorubicin, and cisplatin in advanced or metastatic bladder cancer: results of a large, randomized, multinational, multicenter, phase III study. J. Clin. Oncol. 18, 3068–3077 (2000).

    Article  CAS  Google Scholar 

  7. Bellmunt, J. et al. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N. Engl. J. Med. 376, 1015–1026 (2017).

    Article  CAS  Google Scholar 

  8. Jones, R. J. et al. Randomized phase II study investigating pazopanib versus weekly paclitaxel in relapsed or progressive urothelial cancer. J. Clin. Oncol. 35, 1770–1777 (2017).

    Article  CAS  Google Scholar 

  9. Loriot, Y. et al. Erdafitinib in locally advanced or metastatic urothelial carcinoma. N. Engl. J. Med. 381, 338–348 (2019).

    Article  CAS  Google Scholar 

  10. Wagle, N. et al. Activating mTOR mutations in a patient with an extraordinary response on a phase I trial of everolimus and pazopanib. Cancer Discov. 4, 546–553 (2014).

    Article  Google Scholar 

  11. Necchi, A. et al. Exceptional response to olaparib in BRCA2-altered urothelial carcinoma after PD-L1 inhibitor and chemotherapy failure. Eur. J. Cancer 96, 128–130 (2018).

    Article  CAS  Google Scholar 

  12. Friedlander, M. et al. Pamiparib in combination with tislelizumab in patients with advanced solid tumours: results from the dose-escalation stage of a multicentre, open-label, phase 1a/b trial. Lancet Oncol. 20, 1306–1315 (2019).

    Article  CAS  Google Scholar 

  13. Bellmunt, J. et al. Prognostic factors in patients with advanced transitional cell carcinoma of the urothelial tract experiencing treatment failure with platinum-containing regimens. J. Clin. Oncol. 28, 1850–1855 (2010).

    Article  Google Scholar 

  14. Massard, C. et al. Safety and efficacy of durvalumab (MEDI4736), an anti-programmed cell death ligand-1 immune checkpoint inhibitor, in patients with advanced urothelial bladder cancer. J. Clin. Oncol. 34, 3119–3125 (2016).

    Article  CAS  Google Scholar 

  15. Bellmunt, J., Powles, T. & Vogelzang, N. J. A review on the evolution of PD-1/PD-L1 immunotherapy for bladder cancer: the future is now. Cancer Treat. Rev. 54, 58–67 (2017).

    Article  CAS  Google Scholar 

  16. Zhang, Q. et al. Prognostic and predictive impact of circulating tumor DNA in patients with advanced cancers treated with immune checkpoint blockade. Cancer Discov. 10, 1842–1853 (2020).

  17. Christensen, E. et al. Early detection of metastatic relapse and monitoring of therapeutic efficacy by ultra-deep sequencing of plasma cell-free DNA in patients with urothelial bladder carcinoma. J. Clin. Oncol. 37, 1547–1557 (2019).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  19. Taber, A. et al. Molecular correlates of cisplatin-based chemotherapy response in muscle invasive bladder cancer by integrated multi-omics analysis. Nat. Commun. 11, 4858 (2020).

    Article  CAS  Google Scholar 

  20. Choi, W. et al. Identification of distinct basal and luminal subtypes of muscle-invasive bladder cancer with different sensitivities to frontline chemotherapy. Cancer Cell 25, 152–165 (2014).

    Article  CAS  Google Scholar 

  21. Kamoun, A. et al. A consensus molecular classification of muscle-invasive bladder cancer. Eur. Urol. 77, 420–433 (2020).

    Article  Google Scholar 

  22. Rosenberg, J. E. et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet 387, 1909–1920 (2016).

    Article  CAS  Google Scholar 

  23. Grivas, P. et al. Rucaparib for recurrent, locally advanced, or metastatic urothelial carcinoma (mUC): results from ATLAS, a phase II open-label trial. J. Clin. Oncol. 38, 440–440 (2020).

    Article  Google Scholar 

  24. Update on the Phase III NEPTUNE Trial of Imfinzi Plus Tremelimumab in Stage IV Non-small Cell Lung Cancer (AstraZeneca, 2019); https://www.astrazeneca.com/media-centre/press-releases/2019/update-on-the-phase-iii-neptune-trial-of-imfinzi-plus-tremelimumab-in-stage-iv-non-small-cell-lung-cancer-21082019.html

  25. Powles, T. et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. Lancet 391, 748–757 (2018).

    Article  CAS  Google Scholar 

  26. Powles, T. et al. Clinical efficacy and biomarker analysis of neoadjuvant atezolizumab in operable urothelial carcinoma in the ABACUS trial. Nat. Med. 25, 1706–1714 (2019).

    Article  CAS  Google Scholar 

  27. Pairawan, S. et al. Cell-free circulating tumor DNA variant allele frequency associates with survival in metastatic cancer. Clin. Cancer Res. 26, 1924–1931 (2020).

    Article  CAS  Google Scholar 

  28. Vandekerkhove, G. et al. Plasma ctDNA is a tumor tissue surrogate and enables clinical-genomic stratification of metastatic bladder cancer. Nat. Commun. 12, 184 (2021).

    Article  CAS  Google Scholar 

  29. Xing, P. et al. Co-mutational assessment of circulating tumour DNA (ctDNA) during osimertinib treatment for T790M mutant lung cancer. J. Cell Mol. Med. 23, 6812–6821 (2019).

    Article  CAS  Google Scholar 

  30. Leonetti, A. et al. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br. J. Cancer 121, 725–737 (2019).

    Article  Google Scholar 

  31. Antonescu, C. R. et al. Acquired resistance to imatinib in gastrointestinal stromal tumor occurs through secondary gene mutation. Clin. Cancer Res. 11, 4182–4190 (2005).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the patients and their families who gave their time and commitment to participate in this study; the CRUK Experimental Cancer Medicine Centre; and staff and investigators at participating sites, including the following. Canada: M. Sawyer, University of Alberta—Cross Cancer Institute; S. S. Sridhar, Princess Margaret Cancer Centre, Toronto; C. Ferrario, Jewish General Hospital, Montreal. France: A. Fléchon, Centre Léon Bérard, Lyon; G. Gravis, Institute Paoli-Calmettes, Marseille; F. Joly, Centre Francois Baclesse, Caen; L. Mourey, Institut Claudius Regaud, Toulouse; A. Ravaud, Bordeaux University Hospital; F. Rolland, Institut de Cancerologie de l’Ouest, Saint-Herblain. Spain: J. Carles, Vall D’Hebron Institute of Oncology, Barcelona; J. P. Maroto, Hospital de la Santa Creu i San Pau, Barcelona; J. P. Vásquez, Hospital Clinico San Carlos, Madrid; A. Rodriquez-Vida, IMIM Hospital del Mar Medical Research Instutute, Barcelona. UK: H.-T. Arkenau, Sarah Cannon Research Institute, London; S. Chowdhury, Guys and St Thomas’ Medical School, London; N. Cook, The Christie NHS Foundation Trust, Manchester (Research at the Christie NHS Foundation Trust was supported by the NIHR Manchester Clinical Research Facility and Manchester Experimental Cancer Medicine Centre award); S. Crabb, University of Southampton; R. Jones, University of Glasgow, Beatson West of Scotland Cancer Centre, Glasgow; T. Powles, Barts Cancer Institute, London. USA: A. V. Balar, Perlmutter Cancer Center, NYU Langone Health, New York; J. Bendell, Tennessee Oncology, Nashville; A. Drakaki, Ronald Reagan UCLA Medical Center, Hematology Oncology, Santa Monica; P. Grivas, University of Washington, Seattle; M. Galsky, Icahn School of Medicine at Mount Sinai, New York; N. Hahn, Johns Hopkins University School of Medicine, Baltimore; E. Lim, New York Presbyterian, Columbia University Irving Medical Centre, New York; D. Petrylak, Yale School of Medicine, New Haven; J. Reeves Jr., Florida Cancer Specialists and Research Institute; B. Rini and P. Grivas, Cleveland Clinic, Cleveland; P. Van Veldhuizen, HCA Midwest, Kansas City. We thank the staff at Sarah Cannon Development Innovations and AstraZeneca who supported this clinical study. Medical writing support was provided by S. Hurrell (Bioscript Medical) and was funded by AstraZeneca. We thank A. Reddy (AstraZeneca) for support with data analysis.

Author information

Authors and Affiliations

Authors

Contributions

T.P., S.C., D.P., A.J.B., P.G., D.H., R. Mather, R. McEwen and D.L. contributed to the conception and design of the study. T.P., A.F., D.P., S.S.S., M.D.G., R.J., I.K. and R. Mather acquired the data. T.P., S.C., D.P., A.J.B. S.S.S., M.G., P.G., R.J., D.H., I.K., R. Mather, R. McEwen, F.M. and D.L. contributed to data analysis and interpretation. T.P., S.C., J. Carles, A.F., P.M., D.P., G.G., F.R., N.C., A.J.B., S.S.S., M.G., P.G., A.R., R.J., J. Cosaert and R. Mather contributed to clinical management, patient recruitment and data collection. T.P., A.F., D.P., D.H. and R. Mather contributed administrative, technical or material support. T.P., S.C., D.P., R. Mather, R. McEwen and F.M. contributed to the drafting of the manuscript. T.P., S.C., J. Carles, A.F, P.M., D.P., N.C., A.J.B., S.S.S., M.G., P.G., A.R., R.J., J. Cosaert, D.H., I.K. and D.L. critically revised the manuscript for intellectual content. All authors reviewed and approved the final version of the manuscript.

Corresponding author

Correspondence to Thomas Powles.

Ethics declarations

Competing interests

T.P. has received research funding from AstraZeneca, Astellas, Bristol-Myers Squibb, Roche and Merck; and received honoraria for lectures or advisory boards from AstraZeneca, Astellas, Bristol-Myers Squibb, Roche, Merck, Johnson & Johnson, Ipsen, Exelixis, Pfizer, Novartis and Seattle Genetics. D.C. is a full-time employee at AstraZeneca and owns stocks/shares in AstraZeneca. S.C. has held advisory roles for Astellas Pharma, Bayer, Beigene, Clovis Oncology, Janssen-Cilag, and Pfizer; participated in speakers’ bureaux for Pfizer; received honoraria from Clovis Oncology and Novartis; received research funding from Clovis Oncology and Sanofi/Aventis; and received travel expenses from Beigene and Clovis Oncology. G.G. has received travel expenses for symposia from Bristol-Myers Squibb, Sanofi, Astellas, Ipsen, Janssen and Pfizer. F.J. has provided consultancy for Roche, Ipsen, AstraZeneca, Janssen, Tesaro, Bristol-Myers Squibb, Pfizer, Novartis and Sanofi, Astellas. F.J. has received travel/accommodation expenses from Roche, Ipsen, AstraZeneca, Janssen, Tesaro and Bristol-Myers Squibb and has received funding (institution) from Astellas. J. Carles has received research funding from AB Science, Aragon Pharmaceuticals, Arog Pharmaceuticals, Inc., Astellas Pharma, AstraZeneca AB, Aveo Pharmaceuticals, Bayer AG, Blueprint Medicines, BN Immunotherapeutics, Boehringer Ingelheim España, Bristol-Myers Squibb, Clovis Oncology, Cougar Biotechnology, Deciphera Pharmaceuticlas, Exelixis, Hoffmann-La Roche, Genentech, GlaxoSmithKline, Incyte Corporations, Janssen-Cilag, Karyopharm Therapeutics, Laboratoires Leurquin Mediolanum, Lilly, MedImmune, Millennium Pharmaceuticals, Nanobiotix, Novartis Farmaceutica, Pfizer, Puma Biotechnology, Sanofi-Aventis, SFJ Pharma and Teva; has participated in advisory boards for Bayer, Johnson & Johnson, Bristol-Myers Squibb, Astellas, Pfizer, Sanofi, MSD Oncology, Roche and AstraZeneca; has participated in speakers’ bureaux for Bayer, Johnson & Johnson, Asofarma and Astellas; and received travel expenses from Bristol-Myers Squibb, Ipsen, Roche and AstraZeneca. A.F. has received honoraria and travel expenses from AstraZeneca, MSD, Roche, Janssen and Astellas. P.M. declares no competing interests. D.P. consults for Ada Cap (Advanced Accelerator Applications), Amgen, Astellas, AstraZeneca, Bayer, Bicycle Therapeutics, Boehringer Ingelheim, Bristol-Myers Squibb, Clovis Oncology, Eli Lilly, Exelixis, Incyte, Janssen, Mirati, Monopteros, Pfizer, Pharmacyclics, Roche, Seattle Genetics and Urogen; has received research funding from Ada Cap, Agensys, Astellas, AstraZeneca, Bayer, BioXcel Therapeutics, Bristol-Myers Squibb, Clovis Oncology, Eisai, Eli Lilly, Endocyte, Genentech, Innocrin, MedImmune, Medivation, Merck, Mirati, Novartis, Pfizer, Progenics and Replimune; and is a stockholder in Bellicum. F.R. has received honoraria from Merck KGaA and MSD. N.C. has received research funding from RedX Pharmaceuticals, Tarveda, AstraZeneca, Roche, Novartis, Eisai, Boehringer Ingelheim and Taiho; has participated in advisory boards for RedX Pharmaceuticals; and has been an advisor (unpaid) to Roche Pharmaceuticals. Research at the Christie NHS Foundation Trust was supported by the NIHR Manchester Clinical Research Facility and Manchester Experimental Cancer Medicine Centre award. A.B. has received research funding from Genentech, Merck, AstraZeneca, MedImmune, Nektar, Seattle Genetics and Immunomedics; has held a consultancy or advisory role for Genentech, Incyte, Merck, Pfizer, AstraZeneca, MedImmune, Nektar and Seattle Genetics; has had speaker engagements for Genentech, Merck, AstraZeneca and MedImmune; and has participated in steering groups and advisory boards for Merck. S.S.S. has consulted for or served on advisory boards for AstraZeneca, Pfizer, Roche, Merck, Bristol-Myers Squibb, Astellas, Janssen and Bayer. M.G. has held a consultancy or advisory role for Aileron Therapeutics and is a stockholder in Rappta Therapeutics. P.G. has consulted for AstraZeneca, Bayer, Biocept, Bristol-Myers Squibb, Clovis Oncology, Driver, EMD Serono, Exelixis, Foundation Medicine, Genentech/Roche, Genzyme, Heron Therapeutics, Janssen, Merck, Mirati Therapeutics, Pfizer, Seattle Genetics, QED Therapeutics and GlaxoSmithKline; has delivered educational programs to GlaxoSmithKline; and has received research funding from AstraZeneca, Bayer, Genentech/Roche, Merck, Mirati Therapeutics, Oncogenex, Pfizer, Clovis Oncology, Bavarian Nordic, Immunomedics, Debiopharm and Bristol-Myers Squibb. A.R. has received research funding from Pfizer and Merck GA; and has received honoraria and participated in advisory boards for Pfizer, Merck GA, Bristol-Myers Squibb, AstraZeneca, Roche, Novartis, MSD and Ipsen. R.J. has received research funding from AstraZeneca, MSD, Merck Serono, Pfizer, Bristol-Myers Squibb, Roche, Janssen and Astellas; has received honoraria for speaking and advisory boards from AstraZeneca, MSD, Merck Serono, Pfizer, Bristol-Myers Squibb, Janssen and Astellas; and has written for the educational company Mirrors of Medicine (noncompensated). J. Cosaert is an AstraZeneca employee. D.H., I.K., R. Mather and R. McEwen are AstraZeneca employees and stockholders. F.M. is a contract employee of AstraZeneca and shareholder in StatProcess and Health Data Process. D.L. is a former employee of AstraZeneca (as Senior Director Physician on this study), consults for AstraZeneca and has received research funding from Decision Science.

Additional information

Peer review information Nature Medicine thanks David McConkey, Sumanta Pal and the other, anonymous reviewers for their contribution to the peer review of this work. Javier Carmona was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Consort diagram for screened and enrolled participants.

Consort diagram for screened and enrolled participants. Enrolment periods: AZD4547 monotherapy and AZD4547 + durvalumab, Oct 2016 to Jun 2018; olaparib + durvalumab (selected), Feb 2017 to April 2018; olaparib + durvalumab (unselected), Aug 2018 to Mar 2019; vistusertib + durvalumab, Apr 2017 to Jan 2018; durvalumab monotherapy, Oct 2016 to May 2017. R, randomized.

Extended Data Fig. 2 Most frequent DNA alterations from archived tissue.

Common alterations: Mutations: 65% TERT promoter, 59% TP53, 36% KMT2D, 27% KDM6A and 23% KMT2C. Copy number alterations: 31% CDKN2A loss, 31% CDKN2B loss, 12% MDM2 amplification, 12% FRS2 amplification, 11% CCND1 amplification.

Extended Data Fig. 3

Molecular classification of tumours derived from RNA sequencing data.

Extended Data Fig. 4 TGE signature (IFNG, CXCL9, CD274) comparison (n = 22 and n = 18 subjects in AZD4547 and durvalumab arms, respectively).

Each boxplot displays the median, first and third quartiles (lower and upper hinges), the largest value smaller and the smallest value larger than 1.5 interquartile (upper and lower whiskers).

Extended Data Fig. 5 OS according to tumor TMB.

TMB < 10 mutations/Mb was associated with numerically shorter OS when combining the arms irrespective of treatment (HR 1.924 [0.953; 3.884] from Cox model) Logrank test, P = 0.063. HR, hazard ratio; OS, overall survival; TMB, tumor mutational burden.

Extended Data Fig. 6 PFS according to tumor TMB.

TMB < 10 mutations/Mb was associated with shorter PFS when combining the arms irrespective of treatment (HR 1.904 [1.178; 3.078]) Logrank test, P = 0.007. Analysis performed: Kaplan Meier analysis according to the TMB at baseline and Cox model for HR. HR, hazard ratio; PFS, progression-free survival; TMB, tumor mutational burden.

Extended Data Fig. 7 OS according to blood TMB.

TMB ≤ 16 mutations/Mb was associated with similar OS when combining the arms irrespective of treatment (HR 1.21 [0.69–2.13] from Cox model) Logrank test: p = 0.50. HR, hazard ratio; OS, overall survival; TMB, tumor mutational burden.

Extended Data Fig. 8 PFS according to blood TMB.

TMB ≤ 16 mutations/Mb was associated with shorter PFS when combining the arms irrespective of treatment (HR 1.60 [1.05; 2.42]) Logrank test, p = 0.0025. Analysis performed: Kaplan Meier analysis according to the TMB at baseline and Cox model for HR. HR, hazard ratio; PFS, progression-free survival; TMB, tumor mutational burden.

Extended Data Fig. 9 Oncoprint for ctDNA (Guardant Health) n = 111 from baseline samples (patients receiving treatment).

bTMB, blood-derived tumor mutational burden; ctDNA, circulating tumor deoxyribonucleic acid; DDRm, DNA damage repair mutation.

Extended Data Fig. 10 Correlation between expression of mutations and radiological response.

ctDNA, circulating tumor deoxyribonucleic acid; PD, progressive disease; PR, partial response; SD, stable disease.

Supplementary information

Supplementary Information

Supplementary Figs. 1 and 2 and Supplementary Tables 1–4.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Powles, T., Carroll, D., Chowdhury, S. et al. An adaptive, biomarker-directed platform study of durvalumab in combination with targeted therapies in advanced urothelial cancer. Nat Med 27, 793–801 (2021). https://doi.org/10.1038/s41591-021-01317-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-021-01317-6

This article is cited by

Search

Quick links

Nature Briefing: Cancer

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

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