Intratumoural evolutionary landscape of high-risk prostate cancer: the PROGENY study of genomic and immune parameters

M Linch, G Goh, C Hiley, Y Shanmugabavan, N McGranahan, A Rowan, Y N S Wong, H King, A Furness, A Freeman, J Linares, A Akarca, J Herrero, R Rosenthal, N Harder, G Schmidt, G A Wilson, N J Birkbak, R Mitter, S Dentro, P Cathcart, M Arya, E Johnston, R Scott, M Hung, M Emberton, G Attard, Z Szallasi, S Punwani, S A Quezada, T Marafioti, M Gerlinger, H U Ahmed, C Swanton, M Linch, G Goh, C Hiley, Y Shanmugabavan, N McGranahan, A Rowan, Y N S Wong, H King, A Furness, A Freeman, J Linares, A Akarca, J Herrero, R Rosenthal, N Harder, G Schmidt, G A Wilson, N J Birkbak, R Mitter, S Dentro, P Cathcart, M Arya, E Johnston, R Scott, M Hung, M Emberton, G Attard, Z Szallasi, S Punwani, S A Quezada, T Marafioti, M Gerlinger, H U Ahmed, C Swanton

Abstract

Background: Intratumoural heterogeneity (ITH) is well recognised in prostate cancer (PC), but its role in high-risk disease is uncertain. A prospective, single-arm, translational study using targeted multiregion prostate biopsies was carried out to study genomic and T-cell ITH in clinically high-risk PC aiming to identify drivers and potential therapeutic strategies.

Patients and methods: Forty-nine men with elevated prostate-specific antigen and multiparametric-magnetic resonance imaging detected PC underwent image-guided multiregion transperineal biopsy. Seventy-nine tumour regions from 25 patients with PC underwent sequencing, analysis of mutations, copy number and neoepitopes combined with tumour infiltrating T-cell subset quantification.

Results: We demonstrated extensive somatic nucleotide variation and somatic copy number alteration heterogeneity in high-risk PC. Overall, the mutational burden was low (0.93/Megabase), but two patients had hypermutation, with loss of mismatch repair (MMR) proteins, MSH2 and MSH6. Somatic copy number alteration burden was higher in patients with metastatic hormone-naive PC (mHNPC) than in those with high-risk localised PC (hrlPC), independent of Gleason grade. Mutations were rarely ubiquitous and mutational frequencies were similar for mHNPC and hrlPC patients. Enrichment of focal 3q26.2 and 3q21.3, regions containing putative metastasis drivers, was seen in mHNPC patients. We found evidence of parallel evolution with three separate clones containing activating mutations of β-catenin in a single patient. We demonstrated extensive intratumoural and intertumoural T-cell heterogeneity and high inflammatory infiltrate in the MMR-deficient (MMRD) patients and the patient with parallel evolution of β-catenin. Analysis of all patients with activating Wnt/β-catenin mutations demonstrated a low CD8+/FOXP3+ ratio, a potential surrogate marker of immune evasion.

Conclusions: The PROGENY (PROstate cancer GENomic heterogeneitY) study provides a diagnostic platform suitable for studying tumour ITH. Genetic aberrations in clinically high-risk PC are associated with altered patterns of immune infiltrate in tumours. Activating mutations of Wnt/β-catenin signalling pathway or MMRD could be considered as potential biomarkers for immunomodulation therapies.

Clinical trials.gov identifier: NCT02022371.

Keywords: intratumoural heterogeneity; mismatch repair; neoepitopes; prostate cancer; tumour infiltrating lymphocytes; wnt signalling.

© The Author 2017. Published by Oxford University Press on behalf of the European Society for Medical Oncology.

Figures

Figure 1.
Figure 1.
Intratumoural heterogeneity in prostate cancer at the somatic nucleotide variation (SNV) and somatic copy number alteration (SCNA) levels. (A) Number of somatic exonic mutations identified in each tumour region, fraction of SNVs and SCNAs that were ubiquitous (present in every tumour region of a given patient) (blue), shared (present in more than one tumour region, but not all) (light orange) or private (present in only one tumour region) (dark orange). Data tracks below indicate if patient was metastatic on presentation (red), Gleason grade (shades of green), level of tumoural inflammatory infiltrate (shades of brown), and if the tumour had undergone whole-genome doubling (purple, triangle indicating heterogeneous genome doubling). (B) Scatterplot showing correlation between degree of SNV and SCNA heterogeneity. (C and D) Boxplots comparing the fraction of genome affected by SCNA and SNV mutational burden in metastatic hormone naive prostate cancer (mHNPC) versus high-risk localised prostate cancer (hrlPC).
Figure 2.
Figure 2.
Clonal and subclonal driver events in prostate cancer. List of driver genes previously reported as significantly mutated in primary prostate cancer (blue), metastatic castrate-resistant prostate cancer (mCRPC) (grey), or both (black). Ubiquitous ETS fusion (purple), homozygous loss (dark blue), heterozygous deletion (blue), amplification (red) in each tumour is depicted by a coloured square, and heterozygous events are indicated with a triangle. Nonsynonymous mutations are depicted as smaller squares, whether missense (green), frameshift (yellow) or nonsense (dark blue). Clonal and subclonal mutations are indicated by a purple and orange outline, respectively. Known recurrent mutations in TP53, PIK3CA, CTNNB1 and BRAF are indicated with a red star. The barplots on the right are an aggregate of clonal/ubiquitous or subclonal/heterogenous events in each gene across all samples. Metastatic hormone naive prostate cancer (mHNPC); high-risk localised prostate cancer (hrlPC).
Figure 3.
Figure 3.
Recurrent somatic copy number alterations (SCNAs) in prostate cancer. (A) An overview of the SCNA landscape across all 25 tumours: fraction of cohort (y-axis) with ubiquitous gains (red), heterogeneous gains (pink), ubiquitous loss (dark blue) and heterogeneous loss (light blue) are shaded across the genome (x-axis). (B) Frequencies of occurrence of previously identified GISTIC focal and arm-level SCNAs across all tumours, metastatic on presentation (mHNPC) and non-metastatic on presentation (hrlPC) tumours. Shades of colours as in A.
Figure 4.
Figure 4.
Parallel evolution in Wnt pathway in PR0139 and association with CD8+/FOXP3+ ratio across 15 tumours. (A) Left, fraction of cancer cells in sequenced tumour regions R1, R2 and R3 harbouring different CTNNB1 mutations, p.S33P (pink), p.T41A (blue) and p.S33C (orange). Right, schematic showing different compositions of subclones in each sequenced tumour region, colours correspond to left panel. (B) Phylogenetic tree showing evolutionary history of PR0139 and acquisition of various driver mutations. Relative sizes of circles correspond to number of SNV mutations in that mutational cluster.
Figure 5.
Figure 5.
T-cell heterogeneity in prostate tumours. (A) Manual quantification of inflammatory infiltrate. Mean is represented by horizontal lines, box and whiskers show the 95% confidence interval and range, respectively. The dotted line marks the threshold for high inflammatory infiltrate. (B) Multiplex immunohistochemistry (IHC) analysis of four different prostate core biopsies (R1–4) from a patient, PR0123, showing heterogeneity in T-cell infiltration. CD8 staining in red, CD4 in brown and FoxP3 in blue. (C) Boxplot comparing CD8+/FOXP3+ ratios between tumours with and without somatic activation of Wnt pathway (gain-of-function mutation in CTNNB1, amplification in RSPO2, loss in APC, RNF43 and ZNRF3) across 15 tumours with digital image analysis.

References

    1. World Health Organization. Global Health Observatory (GHO) data: cancer mortality and morbidity 2016.
    1. Hamdy FC, Donovan JL, Lane JA. et al. 10-year outcomes after monitoring, surgery, or radiotherapy for localized prostate cancer. N Engl J Med 2016; 375: 1415–1424.
    1. Gnanapragasam VJ, Lophatananon A, Wright KA. et al. Improving clinical risk stratification at diagnosis in primary prostate cancer: a prognostic modelling study. PLoS Med 2016; 13: e1002063..
    1. Arora R, Koch MO, Eble JN. et al. Heterogeneity of Gleason grade in multifocal adenocarcinoma of the prostate. Cancer 2004; 100: 2362–2366.
    1. Cooper CS, Eeles R, Wedge DC. 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 2015.
    1. Boutros PC, Fraser M, Harding NJ. et al. Spatial genomic heterogeneity within localized, multifocal prostate cancer. Nat Genet 2015; 47: 736–745.
    1. Ahmed HU, El-Shater Bosaily A, Brown LC. et al. Diagnostic accuracy of multi-parametric MRI and TRUS biopsy in prostate cancer (PROMIS): a paired validating confirmatory study. Lancet 2017; 389: 815–822.
    1. Morris LG, Riaz N, Desrichard A. et al. Pan-cancer analysis of intratumor heterogeneity as a prognostic determinant of survival. Oncotarget 2016; 7: 10051–10063.
    1. Lindberg J, Klevebring D, Liu W. et al. Exome sequencing of prostate cancer supports the hypothesis of independent tumour origins. Eur Urol.2013; 63: 347–353.
    1. Wei L, Wang J, Lampert E. et al. Intratumoral and intertumoral genomic heterogeneity of multifocal localized prostate cancer impacts molecular classifications and genomic prognosticators. Eur Urol 2017; 71: 183–192.
    1. Davidsson S, Ohlson AL, Andersson SO. et al. CD4 helper T cells, CD8 cytotoxic T cells, and FOXP3(+) regulatory T cells with respect to lethal prostate cancer. Mod Pathol 2013; 26: 448–455.
    1. Karja V, Aaltomaa S, Lipponen P. et al. Tumour-infiltrating lymphocytes: a prognostic factor of PSA-free survival in patients with local prostate carcinoma treated by radical prostatectomy. Anticancer Res 2005; 25: 4435–4438.
    1. McArdle PA, Canna K, McMillan DC. et al. The relationship between T-lymphocyte subset infiltration and survival in patients with prostate cancer. Br J Cancer 2004; 91: 541–543.
    1. Akarca AU, Shende VH, Ramsay AD. et al. BRAF V600E mutation-specific antibody, a sensitive diagnostic marker revealing minimal residual disease in hairy cell leukaemia. Br J Haematol 2013; 162: 848–851.
    1. Cancer Genome Atlas Research Network. The molecular taxonomy of primary prostate cancer. Cell 2015; 163: 1011–1025.
    1. Andor N, Graham TA, Jansen M. et al. Pan-cancer analysis of the extent and consequences of intratumor heterogeneity. Nat Med 2016; 22: 105–113.
    1. Robinson D, Van Allen EM, Wu YM. et al. Integrative clinical genomics of advanced prostate cancer. Cell 2015; 161: 1215–1228.
    1. Demichelis F, Rubin MA.. TMPRSS2-ETS fusion prostate cancer: biological and clinical implications. J Clin Pathol 2007; 60: 1185–1186.
    1. Baca SC, Prandi D, Lawrence MS. et al. Punctuated evolution of prostate cancer genomes. Cell 2013; 153: 666–677.
    1. Snyder A, Makarov V, Merghoub T. et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 2014; 371: 2189–2199.
    1. Rizvi NA, Hellmann MD, Snyder A. et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015; 348: 124–128.
    1. van Rooij N, van Buuren MM, Philips D. et al. Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J Clin Oncol 2013; 31: e439–e442.
    1. Spranger S, Bao R, Gajewski TF.. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature 2015; 523: 231–235.
    1. Preston CC, Maurer MJ, Oberg AL. et al. The ratios of CD8+ T cells to CD4+CD25+ FOXP3+ and FOXP3- T cells correlate with poor clinical outcome in human serous ovarian cancer. PLoS One 2013; 8: e80063..
    1. Asano Y, Kashiwagi S, Goto W. et al. Tumour-infiltrating CD8 to FOXP3 lymphocyte ratio in predicting treatment responses to neoadjuvant chemotherapy of aggressive breast cancer. Br J Surg 2016; 103: 845–854.
    1. Kirk R. Risk factors: CD8+:FOXP3+ cell ratio is a novel survival marker for colorectal cancer. Nat Rev Clin Oncol 2010; 7: 299–299.
    1. Giannakis M, Hodis E, Jasmine Mu X. et al. RNF43 is frequently mutated in colorectal and endometrial cancers. Nat Genet 2014; 46: 1264–1266.
    1. Hieronymus H, Schultz N, Gopalan A. et al. Copy number alteration burden predicts prostate cancer relapse. Proc Natl Acad Sci USA 2014; 111: 11139–11144.
    1. Gundem G, Van Loo P, Kremeyer B. et al. The evolutionary history of lethal metastatic prostate cancer. Nature 2015; 520: 353–357.
    1. Ishiguro H, Akimoto K, Nagashima Y. et al. aPKClambda/iota promotes growth of prostate cancer cells in an autocrine manner through transcriptional activation of interleukin-6. Proc Natl Acad Sci U S A 2009; 106: 16369–16374.
    1. Gerlinger M, Rowan AJ, Horswell S. et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 2012; 366: 883–892.
    1. Kypta RM, Waxman J.. Wnt/beta-catenin signalling in prostate cancer. Nat Rev Urol 2012; 9: 418–428.
    1. Dominguez-Valentin M, Joost P, Therkildsen C. et al. Frequent mismatch-repair defects link prostate cancer to Lynch syndrome. BMC Urol 2016; 16: 15..
    1. Pritchard CC, Morrissey C, Kumar A. et al. Complex MSH2 and MSH6 mutations in hypermutated microsatellite unstable advanced prostate cancer. Nat Comms 2014; 5: 4988.
    1. Le DT, Uram JN, Wang H. et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 2015; 372: 2509–2520.
    1. Graff JN, Alumkal JJ, Drake CG. et al. Early evidence of anti-PD-1 activity in enzalutamide-resistant prostate cancer. Oncotarget 2016; 7: 52810–52817.

Source: PubMed

Подписаться