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:

Multiregion sampling of de novo metastatic prostate cancer reveals complex polyclonality and augments clinical genotyping

Abstract

De novo metastatic prostate cancer is highly aggressive, but the paucity of routinely collected tissue has hindered genomic stratification and precision oncology. Here, we leveraged a rare study of surgical intervention in 43 de novo metastatic prostate cancers to assess somatic genotypes across 607 synchronous primary and metastatic tissue regions plus circulating tumor DNA. Intra-prostate heterogeneity was pervasive and impacted clinically relevant genes, resulting in discordant genotypes between select primary restricted regions and synchronous metastases. Additional complexity was driven by polyclonal metastatic seeding from phylogenetically related primary populations. When simulating clinical practice relying on a single tissue region, genomic heterogeneity plus variable tumor fraction across samples caused inaccurate genotyping of dominant disease; however, pooling extracted DNA from multiple biopsy cores before sequencing can rescue misassigned somatic genotypes. Our results define the relationship between synchronous treatment-sensitive primary and metastatic lesions in men with de novo metastatic prostate cancer and provide a framework for implementing genomics-guided patient management.

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: Comprehensive clinicogenomic profiling of synchronous primary and metastatic prostate cancer.
Fig. 2: Variable tumor fraction in de novo mCSPC.
Fig. 3: Somatic driver gene landscape of de novo mCSPC via multiregion profiling.
Fig. 4: Intrapatient primary and metastatic genomic heterogeneity.
Fig. 5: Variable tumor-suppressor gene alteration status.
Fig. 6: Phylogenetic reconstruction of local cancer evolution and metastatic dissemination.
Fig. 7: Probability of accurately identifying tumor-suppressor alterations with targeted sequencing of prostate biopsy cores.

Similar content being viewed by others

Data availability

The human reference genome hg38 was downloaded from the University of California, Santa Cruz. We used exon and transcription start site coordinates from the RefSeq Matched Annotation from NCBI and EMBL-EBI (MANE) database. All de-identified sequencing data have been deposited in the European Genome–Phenome Archive database under the accession code EGAS00001006466 and are available under standard controlled release. All other data supporting the findings of this study are available within the article (including its supplementary information and data files) and from the lead contact upon reasonable request. The source data for Figs. 15 and 7 and Extended Data Figs. 13, 5, 6, 9 and 10 are provided as source data files. Source data are provided with this paper.

Code availability

Custom computer code utilized for analysis is available on GitHub at https://github.com/amurth/de_novo_cspc_multifocal_sequencing.

References

  1. Helgstrand, J. T. et al. Trends in incidence and 5-year mortality in men with newly diagnosed, metastatic prostate cancer-A population-based analysis of 2 national cohorts. Cancer 124, 2931–2938 (2018).

    Article  PubMed  Google Scholar 

  2. Hamid, A. A. et al. Metastatic hormone-sensitive prostate cancer: toward an era of adaptive and personalized treatment. Am. Soc. Clin. Oncol. Educ. Book 43, e390166 (2023).

    Article  PubMed  Google Scholar 

  3. Kyriakopoulos, C. E. et al. Chemohormonal therapy in metastatic hormone-sensitive prostate cancer: long-term survival analysis of the randomized phase III E3805 CHAARTED trial. J. Clin. Oncol. 36, 1080–1087 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Gravis, G. et al. Burden of metastatic castrate naive prostate cancer patients, to identify men more likely to benefit from early docetaxel: further analyses of CHAARTED and GETUG-AFU15 studies. Eur. Urol. 73, 847–855 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Vapiwala, N., Hofman, M. S., Murphy, D. G., Williams, S. & Sweeney, C. Strategies for evaluation of novel imaging in prostate cancer: putting the horse back before the cart. J. Clin. Oncol. 37, 765–769 (2019).

    Article  CAS  PubMed  Google Scholar 

  6. Nyquist, M. D. et al. Combined TP53 and RB1 loss promotes prostate cancer resistance to a spectrum of therapeutics and confers vulnerability to replication stress. Cell Rep. 31, 107669 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Ku, S. Y. et al. Rb1 and Trp53 cooperate to suppress prostate cancer lineage plasticity, metastasis, and antiandrogen resistance. Science 355, 78–83 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Annala, M. et al. Circulating tumor DNA genomics correlate with resistance to abiraterone and enzalutamide in prostate cancer. Cancer Discov. 8, 444–457 (2018).

    Article  CAS  PubMed  Google Scholar 

  9. de Bono, J. et al. Olaparib for metastatic castration-resistant prostate cancer. N. Engl. J. Med. 382, 2091–2102 (2020).

    Article  PubMed  Google Scholar 

  10. Abida, W. et al. Rucaparib in men with metastatic castration-resistant prostate cancer harboring a BRCA1 or BRCA2 gene alteration. J. Clin. Oncol. 38, 3763–3772 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Abida, W. et al. Analysis of the prevalence of microsatellite instability in prostate cancer and response to immune checkpoint blockade. JAMA Oncol. 5, 471–478 (2019).

    Article  PubMed  Google Scholar 

  12. Sweeney, C. et al. Ipatasertib plus abiraterone and prednisolone in metastatic castration-resistant prostate cancer (IPATential150): a multicentre, randomised, double-blind, phase 3 trial. Lancet 398, 131–142 (2021).

    Article  CAS  PubMed  Google Scholar 

  13. Cooper, C. S. et al. Analysis of the genetic phylogeny of multifocal prostate cancer identifies multiple independent clonal expansions in neoplastic and morphologically normal prostate tissue. Nat. Genet. 47, 367–372 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Espiritu, S. M. G. et al. The evolutionary landscape of localized prostate cancers drives clinical aggression. Cell 173, 1003–1013 (2018).

    Article  CAS  PubMed  Google Scholar 

  15. Cancer Genome Atlas Research Network. The molecular taxonomy of primary prostate cancer. Cell 163, 1011–1025 (2015).

    Article  Google Scholar 

  16. Schaeffer, E. M. et al. NCCN guidelines insights: prostate cancer, version 1.2023. J. Natl. Compr. Canc. Netw. 20, 1288–1298 (2022).

    PubMed  Google Scholar 

  17. Mateo, J. et al. Genomics of lethal prostate cancer at diagnosis and castration resistance. J. Clin. Invest. 130, 1743–1751 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Stopsack, K. H. et al. Oncogenic genomic alterations, clinical phenotypes, and outcomes in metastatic castration-sensitive prostate cancer. Clin. Cancer Res. 26, 3230–3238 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Wyatt, A. W. et al. Concordance of circulating tumor DNA and matched metastatic tissue biopsy in prostate cancer. J. Natl. Cancer Inst. 109, djx118 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Herberts, C. et al. Deep whole-genome ctDNA chronology of treatment-resistant prostate cancer. Nature 608, 199–208 (2022).

    Article  CAS  PubMed  Google Scholar 

  21. Vandekerkhove, G. et al. Circulating tumor DNA abundance and potential utility in de novo metastatic prostate cancer. Eur. Urol. 75, 667–675 (2019).

    Article  CAS  PubMed  Google Scholar 

  22. Kohli, M. et al. Clinical and genomic insights into circulating tumor DNA-based alterations across the spectrum of metastatic hormone-sensitive and castrate-resistant prostate cancer. eBioMedicine 54, 102728 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Van der Eecken, K. et al. Tissue- and blood-derived genomic biomarkers for metastatic hormone-sensitive prostate cancer: a systematic review. Eur. Urol. Oncol. 4, 914–923 (2021).

    Article  PubMed  Google Scholar 

  24. Sweeney, C. J. et al. Chemohormonal therapy in metastatic hormone-sensitive prostate cancer. N. Engl. J. Med. 373, 737–746 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Fizazi, K. et al. Abiraterone plus prednisone in metastatic, castration-sensitive prostate cancer. N. Engl. J. Med. 377, 352–360 (2017).

    Article  CAS  PubMed  Google Scholar 

  26. Fizazi, K. et al. Abiraterone plus prednisone added to androgen deprivation therapy and docetaxel in de novo metastatic castration-sensitive prostate cancer (PEACE-1): a multicentre, open-label, randomised, phase 3 study with a 2 × 2 factorial design. Lancet 399, 1695–1707 (2022).

    Article  CAS  PubMed  Google Scholar 

  27. Hussain, M. et al. Darolutamide plus androgen-deprivation therapy and docetaxel in metastatic hormone-sensitive prostate cancer by disease volume and risk subgroups in the phase III ARASENS trial. J. Clin. Oncol. 41, 3595–3607 (2023).

    Article  CAS  PubMed  Google Scholar 

  28. Gilson, C. et al. Genomic profiles of de novo high- and low-volume metastatic prostate cancer: results from a 2-stage feasibility and prevalence study in the STAMPEDE trial. JCO Precis. Oncol. 4, 882–897 (2020).

    Article  PubMed  Google Scholar 

  29. Hussain, M. et al. Tumor genomic testing for >4,000 men with metastatic castration-resistant prostate cancer in the phase III trial PROfound (Olaparib). Clin. Cancer Res. 28, 1518–1530 (2022).

    Article  CAS  PubMed  Google Scholar 

  30. Annala, M. et al. Evolution of castration-resistant prostate cancer in ctDNA during sequential androgen receptor pathway inhibition. Clin. Cancer Res. 27, 4610–4623 (2021).

    Article  CAS  PubMed  Google Scholar 

  31. Jayaram, A. et al. Plasma tumor gene conversions after one cycle abiraterone acetate for metastatic castration-resistant prostate cancer: a biomarker analysis of a multicenter international trial. Ann. Oncol. 32, 726–735 (2021).

    Article  CAS  PubMed  Google Scholar 

  32. Annala, M. et al. Cabazitaxel versus abiraterone or enzalutamide in poor prognosis metastatic castration-resistant prostate cancer: a multicentre, randomised, open-label, phase 2 trial. Ann. Oncol. 32, 896–905 (2021).

    Article  CAS  PubMed  Google Scholar 

  33. Mayrhofer, M. et al. Cell-free DNA profiling of metastatic prostate cancer reveals microsatellite instability, structural rearrangements and clonal hematopoiesis. Genome Med. 10, 85 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Bielski, C. M. et al. Genome doubling shapes the evolution and prognosis of advanced cancers. Nat. Genet. 50, 1189–1195 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Robinson, D. et al. Integrative clinical genomics of advanced prostate cancer. Cell 162, 454 (2015).

    Article  CAS  PubMed  Google Scholar 

  36. Black, J. R. M. & McGranahan, N. Genetic and non-genetic clonal diversity in cancer evolution. Nat. Rev. Cancer 21, 379–392 (2021).

    Article  CAS  PubMed  Google Scholar 

  37. Dentro, S. C. et al. Characterizing genetic intra-tumor heterogeneity across 2,658 human cancer genomes. Cell 184, 2239–2254 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Løvf, M., Zhao, S., Axcrona, U. & Johannessen, B. Multifocal primary prostate cancer exhibits high degree of genomic heterogeneity. Eur. Urol. 75, 498–505 (2019).

    Article  PubMed  Google Scholar 

  39. Boysen, G. et al. SPOP-mutated/CHD1-deleted lethal prostate cancer and abiraterone sensitivity. Clin. Cancer Res. 24, 5585–5593 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Swami, U. et al. Association of SPOP mutations with outcomes in men with de novo metastatic castration-sensitive prostate cancer. Eur. Urol. 78, 652–656 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Litchfield, K. et al. Representative sequencing: unbiased sampling of solid tumor tissue. Cell Rep. 31, 107550 (2020).

    Article  CAS  PubMed  Google Scholar 

  42. Emmett, L. et al. The additive diagnostic value of prostate-specific membrane antigen positron emission tomography computed tomography to multiparametric magnetic resonance imaging triage in the diagnosis of prostate cancer (PRIMARY): a prospective multicentre study. Eur. Urol. 80, 682–689 (2021).

  43. Tomlins, S. A. et al. Real-world performance of a comprehensive genomic profiling test optimized for small tumor samples. JCO Precis. Oncol. 5, PO.20.00472 (2021).

    PubMed  PubMed Central  Google Scholar 

  44. Adalsteinsson, V. A. et al. Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat. Commun. 8, 1324 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Hamid, A. A. et al. Compound genomic alterations of TP53, PTEN, and RB1 tumor suppressors in localized and metastatic prostate cancer. Eur. Urol. 76, 89–97 (2019).

    Article  CAS  PubMed  Google Scholar 

  46. Nava Rodrigues, D. et al. RB1 heterogeneity in advanced metastatic castration-resistant prostate cancer. Clin. Cancer Res. 25, 687–697 (2019).

    Article  PubMed  Google Scholar 

  47. van de Haar, J. et al. Limited evolution of the actionable metastatic cancer genome under therapeutic pressure. Nat. Med. 27, 1553–1563 (2021).

    Article  PubMed  Google Scholar 

  48. Clarke, N. W. et al. Abiraterone and olaparib for metastatic castration-resistant prostate cancer. NEJM Evid. 1, EVIDoa2200043 (2022).

  49. Agarwal, N. et al. TALAPRO-2: phase 3 study of talazoparib (TALA) + enzalutamide (ENZA) versus placebo (PBO) + ENZA as first-line (1L) treatment in patients (pts) with metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 41, LBA17 (2023).

    Article  Google Scholar 

  50. Herberts, C. & Wyatt, A. W. Technical and biological constraints on ctDNA-based genotyping. Trends Cancer. 7, 995–1009 (2021).

    Article  CAS  PubMed  Google Scholar 

  51. Deek, M. P. et al. The mutational landscape of metastatic castration-sensitive prostate cancer: the spectrum theory revisited. Eur. Urol. 80, 632–640 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Kumar, A. et al. Substantial interindividual and limited intraindividual genomic diversity among tumors from men with metastatic prostate cancer. Nat. Med. 22, 369–378 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Gundem, G. et al. The evolutionary history of lethal metastatic prostate cancer. Nature 520, 353–357 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Parker, C. C. et al. Radiotherapy to the primary tumour for newly diagnosed, metastatic prostate cancer (STAMPEDE): a randomised controlled phase 3 trial. Lancet 392, 2353–2366 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Lumen, N. et al. The role of cytoreductive radical prostatectomy in the treatment of newly diagnosed low-volume metastatic prostate cancer. results from the local treatment of metastatic prostate cancer (LoMP) registry. Eur. Urol Open Sci. 29, 68–76 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Phillips, R. et al. Outcomes of observation vs stereotactic ablative radiation for oligometastatic prostate cancer: the ORIOLE phase 2 randomized clinical trial. JAMA Oncol 6, 650–659 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Reiter, J. G. et al. Lymph node metastases develop through a wider evolutionary bottleneck than distant metastases. Nat. Genet. 52, 692–700 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Nguyen, B. et al. Genomic characterization of metastatic patterns from prospective clinical sequencing of 25,000 patients. Cell 185, 563–575 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Berglund, E. et al. Spatial maps of prostate cancer transcriptomes reveal an unexplored landscape of heterogeneity. Nat. Commun. 9, 2419 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Mottet, N. et al. EAU-EANM-ESTRO-ESUR-SIOG guidelines on prostate cancer-2020 update. part 1: screening, diagnosis, and local treatment with curative intent. Eur. Urol. 79, 243–262 (2020).

    Article  PubMed  Google Scholar 

  61. Buelens, S. et al. Multicentre, prospective study on local treatment of metastatic prostate cancer (LoMP study). BJU Int. 129, 699–707 (2022).

    Article  CAS  PubMed  Google Scholar 

  62. van Leenders, G. J. L. H. et al. The 2019 International Society of Urological Pathology (ISUP) consensus conference on grading of prostatic carcinoma. Am. J. Surg. Pathol. 44, e87–e99 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Turkbey, B. et al. Prostate imaging reporting and data system version 2.1: 2019 update of prostate imaging reporting and data system version 2. Eur. Urol. 76, 340–351 (2019).

    Article  PubMed  Google Scholar 

  64. Verbeke, S. L. J. et al. A reappraisal of hemangiopericytoma of bone; analysis of cases reclassified as synovial sarcoma and solitary fibrous tumor of bone. Am. J. Surg. Pathol. 34, 777–783 (2010).

    Article  PubMed  Google Scholar 

  65. Roth, A. et al. PyClone: statistical inference of clonal population structure in cancer. Nat. Methods 11, 396–398 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Satas, G., Zaccaria, S., El-Kebir, M. & Raphael, B. J. DeCiFering the elusive cancer cell fraction in tumor heterogeneity and evolution. Cell Systems 12, 1004–1018 (2021).

  67. Niknafs, N., Beleva-Guthrie, V., Naiman, D. Q. & Karchin, R. Subclonal hierarchy inference from somatic mutations: automatic reconstruction of cancer evolutionary trees from multi-region next generation sequencing. PLoS Comput. Biol. 11, e1004416 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Tarabichi, M. et al. A practical guide to cancer subclonal reconstruction from DNA sequencing. Nat. Methods 18, 144–155 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was funded by the Canadian Institutes of Health Research through a Project Grant to A.W.W. and P.O. and Kom op tegen Kanker (Stand up to Cancer—Flemish Cancer Society; P.O.). Other support for the study was provided by the Prostate Cancer Foundation (including via Young Investigator Awards to E.M.K. and N.M.F.) and the Terry Fox Research Institute New Frontiers Program. Part of the collaborative work between A.W.W. and P.O. was enabled by ‘PrIOMiC’, an International Thematic Network, funded by Ghent University and coordinated by ‘Cancer Research Institute Ghent’. No funding sources were involved in the design or execution of the study. We are grateful to all participating patients and their families.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization was the responsibility of N.L., J.V.D., P.O. and A.W.W. Methodology was the responsibility of E.W.W., K.V.d.E., A.J.M., C.H., J.S., N.M.F., E.R., S.V., N.L., J.V.D., M.A., G.V., P.O. and A.W.W. Software was the responsibility of E.W.W., A.J.M., E.M.K., J.S., S.W.S.N., X.E.C., E.R. and M.A. Formal analysis was conducted by E.W.W., K.V.d.E., A.J.M., E.M.K., C.H., J.S., S.W.S.N., X.E.C., N.M.F., E.R., A.M., K.P., S.V., N.L., J.V.D., B.D.L., M.A. and G.V. Investigation was carried out by E.W.W., K.V.d.E., A.J.M., E.M.W., E.S., C.Q.B., G.D., K.B., A.W., S.V., N.L. and J.V.D. Resources were the responsibility of N.L., P.O., J.S., M.A. and A.W.W. Data curation was carried out by E.W.W., K.V.d.E., A.J.M., E.M.K., S.W.S.N., X.E.C., N.M.F., E.R., E.S., C.Q.B., G.D., K.B., A.W., S.V. and M.A. Writing of the original draft was carried out by E.W.W., K.V.d.E., A.J.M., E.M.K., C.H., J.S., G.V. and A.W.W. Review and editing was conducted by E.W.W., K.V.d.E., A.J.M., E.M.K., C.H., J.S., S.W.S.N., X.E.C., N.M.F., E.R., B.D.L., M.A., G.V., P.O. and A.W.W. Visualization was carried out by E.W.W., K.V.d.E., A.J.M., E.M.K., C.H., J.S., S.W.S.N., X.E.C., N.M.F., E.R. and M.A. Supervision was the responsibility of B.D.L., M.A., G.V., P.O. and A.W.W. Project administration was carried out by N.L., J.V.D., G.V., P.O. and A.W.W. Funding acquisition was the responsibility of N.L., J.V.D., P.O. and A.W.W.

Corresponding author

Correspondence to Alexander W. Wyatt.

Ethics declarations

Competing interests

E.W.W. is an employee at Guardant Health (although only contributed to this article before commencing employment with Guardant Health in April 2023). E.M.K. has consulted or served in an advisory role for Astellas Pharma, Janssen and Ipsen, received travel funding from Astellas Pharma, Pfizer, Ipsen and Roche, received honoraria from Janssen, Ipsen, Astellas Pharma and Research Review and research funding from Astellas Pharma (institutional) and AstraZeneca. M.A. is a shareholder in Fluivia. The laboratory of N.L. has research funding from Bayer (Inst) and Janssen (Inst) and has received travel funding from Ipsen (Inst). P.O. has consulted or served in an advisory role for Janssen-Cilag, Bayer, Astellas Pharma, Curium Pharma, Telix Pharmaceuticals and Novartis and has received travel funding from Ferring. The laboratory of P.O. has received research funding from Varian Medical Systems (Inst) and Bayer (Inst). A.W.W. has served on advisory boards and/or received honoraria from AstraZeneca, Astellas, Bayer, EMD Serono, Janssen, Merck and Pfizer. The laboratory of A.W.W. has a contract research agreement with ESSA Pharma. All other authors declare no competing interests.

Peer review

Peer review information

Nature Cancer thanks the anonymous reviewers 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

Extended Data Fig. 1 Sequencing metrics and tumor fraction estimation.

a. DNA yield, median coverage, and tumor fraction (TF) estimates derived from targeted sequencing. Numbers of independent samples per category annotated in brackets. b. Hierarchical approach for TF estimation. Mutations deemed ineligible for TF estimation in Group 1 (that is mutations not shared across most same-patient samples, and are therefore potentially subclonal) can underestimate TF compared to heterozygous single-nucleotide polymorphism (SNP) allele frequency applied in Group 2 (middle scatter plot). c. Comparison of TF by targeted (left) and whole-exome sequencing (WES; right) sequencing versus pathology tumor cellularity (TC; n = 521). Bars show the fraction of samples with higher pathology-derived than sequencing-derived TF estimates. d. Sequencing-derived TF estimates versus median targeted sequencing coverage (for all tissue samples; n = 523). e. High median coverage of all ISUP grade 5 samples with sequencing TF of 0% indicating that sequencing insufficiencies do not explain low tumor fraction results. f. Sequencing-derived TF estimates for each tissue sample (n are annotated), grouped by patient along the x-axis. ISUP (International Society of Urological Pathology) high and low refer to primary grade ≥4 and <4, respectively). Asterisks denote patients who received preoperative androgen-deprivation therapy (ADT). g. Variant allele frequencies of somatic mutations by targeted sequencing and whole-exome sequencing (WES) are highly correlated. Each dot represents a mutation detected via either sequencing method with jointly sufficient locus-specific depth (n = 234). Mutations with <30x coverage by either method are excluded. h. Coverage log ratio (LR) correlation between 73-gene targeted panel and WES (in samples sequenced with both modalities; n = 152). Each dot (n = 11271) represents a gene shared across panels. i. Few somatic mutations detected solely by WES are within established cancer genes. Of 7757 mutations detected in genes not covered by the targeted sequencing panel, only six were pathogenic coding mutations in key cancer driver genes (moving left to right, each subsequent bar is a subset of the previous bar). cfDNA: cell-free DNA, FFPE: formalin-fixed paraffin-embedded, MLN: metastatic lymph node, PB: prostate biopsy, RP: radical prostatectomy.

Source data

Extended Data Fig. 2 Concurrent metastatic prostate and small-cell lung cancer in ID19.

a. Clinical timeline of cancer diagnoses and treatment for patient ID19 from prostate cancer (PCa) diagnosis until death. b. Variant allele fraction (VAF) of all somatic mutations detected in the cell-free DNA (cfDNA) sample at time of radical prostatectomy and pelvic lymph node (LN) dissection (RP/PLND). Mutations in red were independently detected in prostate tumor tissue. c. Pairwise VAF comparisons between 5 samples with whole-exome sequencing data. Each dot represents the VAF of one mutation independently detected in at least one sample. SCLC: small-cell lung cancer, ADT: androgen-deprivation therapy, CCF: cancer cell fraction, ctDNA: circulating tumor DNA, PLND: pelvic lymph node dissection, PSA: prostate-specific antigen.

Source data

Extended Data Fig. 3 Per-patient aggregate driver gene alteration status.

a. Summary of patient genotypes (n = 43) aggregated from all same-patient samples (n = 607). Variants are shown if they were present in >2 samples (for SNVs/indels and deep deletions/amplifications) or >3 high tumor fraction (<20%) samples (for shallow gains and deletions). ID31 and ID39 did not have sufficient high tumor fraction samples for copy number analysis and ID24 did not have any tumor positive samples for mutation or copy number analysis. Asterisks denote patients who received preoperative androgen-deprivation therapy. b-c. Copy number and tumor fraction model from whole-exome sequencing for patient ID5 with whole-genome doubling (b) and ID23 with diploid genome (c), where the top plot of each panel shows coverage log ratio (left axis) and copy number (right axis), and the bottom plot shows germline heterozygous single-nucleotide polymorphism (SNP) allele frequency (HSAF, left axis) and allelic ratio (right axis). Median coverage log ratio and HSAF for each segment are shown in colored thick bars, where colors indicate copy number and allelic ratio. Green segments did not fit to a specific copy number and allelic ratio. d-e. Comparisons of alteration frequencies between our cohort and public cohorts of localized castration-sensitive prostate cancer (CSPC)15, metastatic CSPC (mCSPC)18, and metastatic castration-resistant prostate cancer (mCRPC)35. Patient numbers and alteration frequencies are identical to Fig. 4a. Top (d): circles show the detection rate across all patients in our cohort with variants detected in >1 sample (for single-nucleotide substitutions/indels) or >2 high tumor fraction (>20%) samples (for copy number variants). Bottom (d): circles show the expected frequency of somatic mutations, amplifications, and homozygous losses in our cohort (using an average of single randomly-selected samples per patient) compared to published cohorts. e. Frequency of biallelic TP53, RB1, and PTEN loss across cohorts. TCGA: The Cancer Genome Atlas, MSKCC: Memorial Sloan Kettering Cancer Center, SU2C: Stand Up 2 Cancer.

Source data

Extended Data Fig. 4 Immunohistochemistry staining of prostate tissue to confirm tumor-suppressor gene loss.

Staining of PTEN and RB1 protein in eight representative tumor tissue samples, plus two positive control samples derived from benign tonsil tissue. A total of 26 tumor tissue regions were tested, and genomic and immunohistochemistry results were concordant in 24 of 26 (see Supplementary Table 10). Images were captured at 20× magnification. All scale bars represent 50 µm. Asterisk denotes the patient who received preoperative androgen-deprivation therapy. Experiments were performed once per sample. RP: radical prostatectomy, LOH: loss of heterozygosity, MLN: metastatic lymph node.

Extended Data Fig. 5 Evidence of genomic heterogeneity across same-patient tumor regions.

a. Private versus shared mutations between the primary and metastatic niche. b. Clonal status of all mutations (All muts), and for the subset that are primary-exclusive (P. Private), metastatic-exclusive (M. Private), or shared. Mutation n are provided at the top of each bar. Mutations detected in multiple same-patient samples were defined as clonal if they had a CCF ≥ 0.8 in any sample. c. Per-gene mutation count, with color denoting mutation heterogeneity (private to primary or metastatic niche, or shared). Only data from patients (n = 25 [targeted], n = 29 [whole-exome sequencing; WES]) with both tumor fraction (TF)-positive primary and metastatic samples are included. Top and bottom rows of (a-c) utilize samples subjected to targeted and WES, respectively. Patient (ID8) with mismatch repair deficient disease was excluded. d. Most mutations detected across same-patient samples with WES data are not shared across all tumor regions. 36 patients with multiple TF-positive samples were included. Patient (ID8) with mismatch repair deficient disease was excluded. In-set stripplot: fraction of mutations in each patient that were detected in all TF-positive samples versus number of same-patient samples with WES data, suggesting that sample availability does not impact the proportion of mutations that are detected in all same-patient WES samples. e. Heatmap showing whole-exome sequencing derived copy number profiles for all samples with ≥30% TF (n = 127). Blue denotes copy losses while red denotes copy gains, both relative to base copy number (four copies in samples with evidence of whole-genome duplication [WGD], two for presumed diploid samples). Same-patient samples and WGD are annotated on the left. Asterisks denote patients who received preoperative ADT.

Source data

Extended Data Fig. 6 Evidence for independent primary prostate tumors in three patients.

See Supplementary Fig. 5 for the four other patients with multiple independent cancers. The prostate schematics show the approximate location of profiled prostate biopsy (PB) cores and radical prostatectomy (RP) regions (no map was available for ID38). Estimated extent of tumor involvement (as determined by histopathological review) is shown, and independent tumors (labeled A, B, and C) are indicated. For select representative (‘model’) samples from each independent tumor, the highest observed International Society of Urological Pathology (ISUP) grade and key genomic alterations are shown. Different patterns of copy number alterations in genes (middle) from targeted sequencing for the model samples; coloring indicates deep deletion (dark blue), shallow deletion (light blue), copy-neutral (black), copy-gain (red) and amplification (dark red). Scatterplots (right) and Pearson correlation of all mutations detected via whole-exome sequencing in select sample pairs depict no overlap between independent tumors. Del: deletion, Mut: mutation. ADT: androgen-deprivation therapy, TF: tumor fraction, UCC: urothelial carcinoma, VAF: variant allele fraction.

Source data

Extended Data Fig. 7 Phylogenetic tree topologies and metastatic dissemination patterns.

Evolutionary tree diagrams for all additional patients not provided in Fig. 6 and/or Extended Data Figure 8. Branch colors correspond to PyClone-identified clones, and to patient-specific matrices shown in Supplementary Fig. 7. Patients without metastatic clones shown had no available tumor fraction positive metastatic sample. a. Patients with evidence of multiple independent primary tumors. Localized independent tumors generally display less branching than metastatic lineages. b. Evolutionary trees of all other de novo mCSPC cases. Del: copy number deletion, mut: mutation (single-nucleotide substitution / small insertion or deletion). Equally parsimonious trees for ID17, ID18, ID29, and ID4 are shown in Supplementary Fig. 8e. c. Results for ID8 demonstrating heterogeneous mismatch repair defective status and extensive phylogenetic branching. Each shape (circle = primary, diamond = metastasis) is a Pyclone-identified clone assignment for each sample (n = 12), where shape size represents median cancer cell fraction of mutations within the cluster. MSH2/6 status is shown on the left. Mutations per megabase (Mut/Mb) of each sample are shown on the right and cluster mutation count is shown above. Colors are assigned to the truncal clone and phylogenetically distinct branches. d. Output from PyClone and SCHISM, colored by distinct branches, for ID8. Number of mutations per branch are annotated (branch length does not reflect mutation count). Each colored branch has a unique MSH2/6 status and Mut/Mb in associated samples. All samples with exclusively pink populations, including metastatic lymph nodes, are notably devoid of hypermutation and have intact MSH6, while the two other clusters (yellow and green) exhibit hypermutation and losses of both MSH2 and MSH6. The green clone likely contains a second hit not detectable by WES (for example intronic truncating rearrangement). All three populations colonized the metastatic niche. LR: log ratio, MLN: metastatic lymph node, PB: prostate biopsy, RP: radical prostatectomy, WDG: whole-genome duplication.

Extended Data Fig. 8 Parsimonious migration models depicting intersample clonal seeding.

Plausible clonal dissemination patterns between samples of all patients included in Fig. 6 or patients with multiple metastatic samples (total patient n = 14). Pie plot proportion is not representative of clonal proportion within samples. Clinically-relevant somatic alterations, whole-genome duplication status, and select clinical characteristics are annotated. Unlabeled nodes represent hypothetical monoclonal samples (that is not directly observed/sequenced) to assist with interpretation (via showing stepwise clonal evolution where possible). ADT: androgen-deprivation therapy, del: copy number deletion, mut: mutation (single-nucleotide substitution / small insertion or deletion), MLN: metastatic lymph node, PB: prostate biopsy, RP: radical prostatectomy.

Extended Data Fig. 9 Concordance between plasma ctDNA and same-patient tumor tissue samples.

a. Oncoprint of all same-patient formalin-fixed paraffin-embedded (FFPE) samples and the cell-free DNA (cfDNA) sample with the highest ctDNA fraction collected prior to androgen-deprivation therapy (ADT) initiation, for the 10 patients with detected ctDNA pre-ADT. Asterisks indicate the patient who received ADT prior to radical prostatectomy. Total sample n = 156 (146 tissue; 10 cfDNA). b. ctDNA fraction and prostate-specific antigen (PSA) levels (ng/mL) of 3 patients with pre- and post-radical prostatectomy cfDNA collections. c. Composite oncoprint of all same-patient FFPE samples and the cfDNA sample with the highest ctDNA sample fraction collected after progression to metastatic castration-resistant prostate cancer (mCRPC), for 8 patients. Asterisks indicate patients who received androgen-deprivation theory (ADT) prior to radical prostatectomy. Total sample n = 129 (121 tissue; 9 cfDNA). CNV: copy number variant, LR: log ratio.

Source data

Extended Data Fig. 10 In vitro pooled and sequenced biopsy material recapitulates the average genotypes of constituent input samples and in silico merged samples.

a. Concordance between somatic mutation variant allele frequency (VAF, left column), gene copy number log ratios (center column), and tumor fraction (right column) between in vitro combined versus mean input sample (black), in vitro versus in silico combined samples (blue), and in silico combined versus mean input sample (green). Note that in vitro log ratio values were normalized using white blood cell DNA (rather than source-matched pooled benign tissue as per the other comparator samples). Seven patients are included and the number of mutations assessed is indicated on each plot. b. Genomic status of TP53, PTEN, and RB1 in primary, in silico combined, and in vitro combined samples. Copy number is corrected for tumor fraction. Heterozygous single-nucleotide polymorphism (SNP) variant allele fraction greater than 60% was considered a heterozygous loss. LOH: loss of heterozygosity.

Source data

Supplementary information

Supplementary Information

Supplementary Note & Supplementary Figs. 1–9.

Reporting Summary

Supplementary Table 1

Supplementary Tables 1–17.

Source data

Source Data Fig. 1

Processed data for figure reproduction.

Source Data Fig. 2

Processed data for figure reproduction.

Source Data Fig. 3

Processed data for figure reproduction.

Source Data Fig. 4

Processed data for figure reproduction.

Source Data Fig. 5

Processed data for figure reproduction.

Source Data Fig. 7

Processed data for figure reproduction.

Source Data Extended Data Fig. 1

Processed data for figure reproduction.

Source Data Extended Data Fig. 2

Processed data for figure reproduction.

Source Data Extended Data Fig. 3

Processed data for figure reproduction.

Source Data Extended Data Fig. 5

Processed data for figure reproduction.

Source Data Extended Data Fig. 6

Processed data for figure reproduction.

Source Data Extended Data Fig. 9

Processed data for figure reproduction.

Source Data Extended Data Fig. 10

Processed data for figure reproduction.

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

Warner, E.W., Van der Eecken, K., Murtha, A.J. et al. Multiregion sampling of de novo metastatic prostate cancer reveals complex polyclonality and augments clinical genotyping. Nat Cancer 5, 114–130 (2024). https://doi.org/10.1038/s43018-023-00692-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-023-00692-y

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