Melanoma-specific MHC-II expression represents a tumour-autonomous phenotype and predicts response to anti-PD-1/PD-L1 therapy

Douglas B Johnson, Monica V Estrada, Roberto Salgado, Violeta Sanchez, Deon B Doxie, Susan R Opalenik, Anna E Vilgelm, Emily Feld, Adam S Johnson, Allison R Greenplate, Melinda E Sanders, Christine M Lovly, Dennie T Frederick, Mark C Kelley, Ann Richmond, Jonathan M Irish, Yu Shyr, Ryan J Sullivan, Igor Puzanov, Jeffrey A Sosman, Justin M Balko, Douglas B Johnson, Monica V Estrada, Roberto Salgado, Violeta Sanchez, Deon B Doxie, Susan R Opalenik, Anna E Vilgelm, Emily Feld, Adam S Johnson, Allison R Greenplate, Melinda E Sanders, Christine M Lovly, Dennie T Frederick, Mark C Kelley, Ann Richmond, Jonathan M Irish, Yu Shyr, Ryan J Sullivan, Igor Puzanov, Jeffrey A Sosman, Justin M Balko

Abstract

Anti-PD-1 therapy yields objective clinical responses in 30-40% of advanced melanoma patients. Since most patients do not respond, predictive biomarkers to guide treatment selection are needed. We hypothesize that MHC-I/II expression is required for tumour antigen presentation and may predict anti-PD-1 therapy response. In this study, across 60 melanoma cell lines, we find bimodal expression patterns of MHC-II, while MHC-I expression was ubiquitous. A unique subset of melanomas are capable of expressing MHC-II under basal or IFNγ-stimulated conditions. Using pathway analysis, we show that MHC-II(+) cell lines demonstrate signatures of 'PD-1 signalling', 'allograft rejection' and 'T-cell receptor signalling', among others. In two independent cohorts of anti-PD-1-treated melanoma patients, MHC-II positivity on tumour cells is associated with therapeutic response, progression-free and overall survival, as well as CD4(+) and CD8(+) tumour infiltrate. MHC-II(+) tumours can be identified by melanoma-specific immunohistochemistry using commercially available antibodies for HLA-DR to improve anti-PD-1 patient selection.

Conflict of interest statement

D.B.J., J.M.B., V.S. and M.E.S. have filed provisionary patent on use of MHC-II to predict response to immunotherapy. The remaining authors declare no competing financial interests.

Figures

Figure 1. A unique subtype of melanoma…
Figure 1. A unique subtype of melanoma expresses MHC-II.
(a) Microarray data from 60 melanoma cell lines in the CCLE were analysed for MHC-I (HLA-A/B/C and MHC-II (HLA-DRA) expression. Bars represent the mean±s.d. P value is the result of the Kolmogorov–Smirnov test comparing the distribution of MHC-I (HLA-A, HLA-B, HLA-C) expression with MHC-II expression (HLA-DRA). *represents the cutoff for defining MHC-II(+). (b) Gene-expression data from HLA-DRA(+) cell lines (Clusters Ia/Ib) were compared with HLA-DRA(−) cell lines (Clusters II and III) by an FDR-corrected row t-test. Significantly altered genes are shown on the y-axis and also listed in Supplementary Data 1. An ad hoc heat map is shown at the top, highlighting classical MHC-II genes. (c) Normalized microarray data were analysed by GSA using the curated Molecular Signatures Database, and the resulting gene set scores are presented as a hierarchical clustered heat map.
Figure 2. Characterization of MHC-II(+) melanoma cell…
Figure 2. Characterization of MHC-II(+) melanoma cell lines.
Melanoma cell lines were treated with IFNγ for 24 h before collection and live-cell staining and flow cytometry analysis for MHC-I/HLA-A/B/C (a), MHC-II/HLA-DR (b) and PD-L1 (c). Bars represent mean±s.e.m. for at least three experiments (d) Representative flow plots from c. (e) Western blot analysis of melanoma cell lines after 24 or 48 h of IFNγ stimulation. (f) Phosphorylation of STAT1 (top row) and STAT5 (bottom row) in melanoma cell lines at 15 min after IFNγ stimulation. Histograms were coloured according to the arcsinh transformed ratio or MFI medians relative to the table minimum value.
Figure 3. MHC-II-positive melanoma cell lines associate…
Figure 3. MHC-II-positive melanoma cell lines associate with NRAS mutations.
(a) HLA-DRA mRNA expression in melanoma cell lines (n=60; one cell line lacked mRNA expression data) from the CCLE compared by genotype. P value (P<0.05) represents result of Tukey's post hoc analysis comparing pan-WT with NRAS-mutant cell lines, following a significant ANOVA (P=0.03) performed among all groups. Bars represent mean±s.e.m. (b) Representative IHC for HLA-DR (brown) and SOX10 (pink) in cases with isolated stromal positivity (top) and with tumour-specific staining (bottom). Both HLA-DR and SOX10 immunostaining is present in all four sections. Scale bar, 50 μm. (c) Analysis of HLA-DR IHC in a melanoma TMA (n=67 evaluable) by genotype. P value represents result of a χ2-test. (d) Overall survival of patients (n=58 evaluable) within the TMA by HLA-DR status (left censored at time of diagnosis). The remaining patient samples were included from outside institutions and follow-up data were not available from those institutions. P value is the result of the log-rank test.
Figure 4. Ex vivo culture of tumours…
Figure 4. Ex vivo culture of tumours derived from anti-PD-1-responding and non-responding patients identifies heterogeneity in interferon response.
(a) Patient tumour blocks stained for HLA-DR (brown) and SOX10 (pink) at low (scale bar, 500 um) and high magnification (scale bar, 200 μm); PT1: anti-PD-1 non-responder and PT2: anti-PD-1 responder. (b) Experimental schema. (c) Schema and images of PDX tissue sections (ex vivo organotypic culture). (d) Western blot analysis of tissue sections cultured in the presence or absence of IFNγ for 24–48 h.
Figure 5. MHC-II(+) melanomas have improved response…
Figure 5. MHC-II(+) melanomas have improved response rates and clinical benefit to PD-1/PD-L1 inhibition.
(a) HLA-DR positivity by IHC plotted versus response to PD-1/PD-L1-targeted therapy in the discovery set (n=30). Responders include partial and complete responders; non-responders include mixed responders and progressive disease patients. Mixed responders (n=3) are noted by a red triangle. P value is the result of the Wilcoxon's rank sum test. (b) HLA-DR positivity by IHC in the validation set (n=23) plotted versus response to PD-1/PD-L1-targeted therapy. P value is the result of the Wilcoxon's rank sum test (c) Representative images of scans from anti-PD-1 therapy-treated MHC-II(+) patients (d) Progression-free survival (left) and overall survival (right) in anti-PD-1/PD-L1-treated patients, stratified by HLA-DR/MHC-II positivity (5% total tumour cells staining on entire tissue section used as cutpoint). Data from both the initial and validation cohorts were included, when available. P value is the result of the log-rank test. (e) Correlation matrix of IHC markers. P values for the Pearson's correlation appear above the diagonal and correlation coefficients (r) appear below the diagonal.

References

    1. Topalian S. L. et al.. Safety, activity and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366, 2443–2454 (2012).
    1. Hamid O. et al.. Safety and tumour responses with lambrolizumab (anti-PD-1) in melanoma. N. Engl. J. Med. 369, 134–144 (2013).
    1. Herbst R. S. et al.. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 515, 563–567 (2014).
    1. Robert C. et al.. Nivolumab in previously untreated melanoma without BRAF mutation. N. Engl. J. Med. 372, 320–330 (2014).
    1. Robert C. et al.. Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. Lancet 384, 1108–1117 (2014).
    1. Robert C. et al.. Pembrolizumab versus ipilimumab in advanced melanoma. N. Engl. J. Med. 372, 2521–2532 (2015).
    1. Rizvi N. A. et al.. Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (CheckMate 063): a phase 2, single-arm trial. Lancet Oncol. 16, 257–265 (2015).
    1. Garon E. B. et al.. Pembrolizumab for the treatment of non-small-cell lung cancer. N. Engl. J. Med. 372, 2018–2028 (2015).
    1. Gettinger S. N. et al.. Overall survival and long-term safety of nivolumab (anti-programmed death 1 antibody, BMS-936558, ONO-4538) in patients with previously treated advanced non-small-cell lung cancer. J. Clin. Oncol. 33, 2004–2012 (2015).
    1. Motzer R. J. et al.. Nivolumab for metastatic renal cell carcinoma: results of a randomized phase II trial. J. Clin. Oncol. 33, 1430–1437 (2014).
    1. Powles T. et al.. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature 515, 558–562 (2014).
    1. Ansell S. M. et al.. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. N. Engl. J. Med. 372, 311–319 (2014).
    1. Nanda R. et al.. in Proceedings of the 2014 San Antonio Breast Cancer Symposium; 9–13 December 2014; San Antonio, TX. Abstract S1-09 .
    1. Seiwert T. Y. et al.. A phase Ib study of MK-3475 in patients with human papillomavirus (HPV)-associated and non-HPV–associated head and neck (H/N) cancer. J. Clin. Oncol. 32, 6011 (2014).
    1. Brahmer J. R. et al.. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumours: safety, clinical activity, pharmacodynamics, and immunologic correlates. J. Clin. Oncol. 28, 3167–3175 (2010).
    1. Snyder A. et al.. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 371, 2189–2199 (2014).
    1. Yadav M. et al.. Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing. Nature 515, 572–576 (2014).
    1. Tumeh P. C. et al.. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 568–571 (2014).
    1. Spranger S., Bao R. & Gajewski T. F. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature 523, 231–235 (2015).
    1. Mellman I., Coukos G. & Dranoff G. Cancer immunotherapy comes of age. Nature 480, 480–489 (2011).
    1. Mikucki M. E. et al.. Non-redundant requirement for CXCR3 signalling during tumoricidal T-cell trafficking across tumour vascular checkpoints. Nat. Commun. 6, 7458 (2015).
    1. Garrido F., Cabrera T. & Aptsiauri N. “Hard” and “soft” lesions underlying the HLA class I alterations in cancer cells: implications for immunotherapy. Int. J. Cancer 127, 249–256 (2010).
    1. Warabi M., Kitagawa M. & Hirokawa K. Loss of MHC class II expression is associated with a decrease of tumour-infiltrating T cells and an increase of metastatic potential of colorectal cancer: immunohistological and histopathological analyses as compared with normal colonic mucosa and adenomas. Pathol. Res. Pract. 196, 807–815 (2000).
    1. Bernsen M. R. et al.. On the biological relevance of MHC class II and B7 expression by tumour cells in melanoma metastases. Br. J. Cancer 88, 424–431 (2003).
    1. Oldford S. A. et al.. Tumour cell expression of HLA-DM associates with a Th1 profile and predicts improved survival in breast carcinoma patients. Int. Immunol. 18, 1591–1602 (2006).
    1. Degenhardt Y. et al.. Distinct MHC gene expression patterns during progression of melanoma. Genes Chromosomes Cancer 49, 144–154 (2010).
    1. Pollack M. S., Heagney S. D., Livingston P. O. & Fogh J. HLA-A, B, C and DR alloantigen expression on forty-six cultured human tumour cell lines. J. Natl Cancer Inst. 66, 1003–1012 (1981).
    1. Barbieri G., Rimini E. & Costa M. A. Effects of human leukocyte antigen (HLA)-DR engagement on melanoma cells. Int. J. Oncol. 38, 1589–1595 (2011).
    1. Colloby P. S., West K. P. & Fletcher A. Is poor prognosis really related to HLA-DR expression by malignant melanoma cells? Histopathology 20, 411–416 (1992).
    1. Chornoguz O., Gapeev A., O'Neill M. C. & Ostrand-Rosenberg S. Major histocompatibility complex class II+ invariant chain negative breast cancer cells present unique peptides that activate tumour-specific T cells from breast cancer patients. Mol. Cell Proteomics 11, 1457–1467 (2012).
    1. Londei M., Lamb J. R., Bottazzo G. F. & Feldmann M. Epithelial cells expressing aberrant MHC class II determinants can present antigen to cloned human T cells. Nature 312, 639–641 (1984).
    1. Meazza R., Comes A., Orengo A. M., Ferrini S. & Accolla R. S. Tumour rejection by gene transfer of the MHC class II transactivator in murine mammary adenocarcinoma cells. Eur. J. Immunol. 33, 1183–1192 (2003).
    1. Rizvi N. A. et al.. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348, 124–128 (2015).
    1. Wellbrock C. et al.. STAT5 contributes to interferon resistance of melanoma cells. Curr. Biol. 15, 1629–1639 (2005).
    1. Lee Y. S., Kim S. H., Cho J. A. & Kim C. W. Introduction of the CIITA gene into tumour cells produces exosomes with enhanced anti-tumour effects. Exp. Mol. Med. 43, 281–290 (2011).
    1. Joseph R. W. et al.. Correlation of NRAS mutations with clinical response to high-dose IL-2 in patients with advanced melanoma. J. Immunother. 35, 66–72 (2012).
    1. Johnson D. B. et al.. Impact of NRAS mutations for patients with advanced melanoma treated with immune therapies. Cancer Immunol. Res. 3, 288–295 (2015).
    1. Loor F. & Kindred B. Differentiation of T-cell precursors in nude mice demonstrated by immunofluorescence of T-cell membrane markers. J. Exp. Med. 138, 1044–1055 (1973).
    1. Rodriguez T. et al.. Patterns of constitutive and IFN-gamma inducible expression of HLA class II molecules in human melanoma cell lines. Immunogenetics 59, 123–133 (2007).
    1. Wolchok J. D. et al.. Nivolumab plus ipilimumab in advanced melanoma. N. Engl. J. Med. 369, 122–133 (2013).
    1. Postow M. A. et al.. Nivolumab and Ipilimumab versus ipilimumab in untreated melanoma. N. Engl. J. Med. 372, 2006–2017 (2015).
    1. Larkin J. et al.. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 373, 23–34 (2015).
    1. Balko J. M. et al.. Profiling of residual breast cancers after neoadjuvant chemotherapy identifies DUSP4 deficiency as a mechanism of drug resistance. Nat. Med. 18, 1052–1059 (2012).
    1. Balko J. M. et al.. Activation of MAPK pathways due to DUSP4 loss promotes cancer stem cell-like phenotypes in basal-like breast cancer. Cancer Res. 73, 6346–6358 (2013).
    1. R Development Core Team. R: A language and environment for statistical computing R Foundation for Statistical Computing: Vienna, Austria, (2015).
    1. Benjamini Y. & Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Series B Stat. Methodol. 57, 289–300 (1995).
    1. Efron B. & Tibshirani R. On testing the significance of sets of genes. Ann. Appl. Stat. 1, 107–129 (2007).
    1. Barretina J. et al.. The cancer cell line encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483, 603–607 (2012).

Source: PubMed

3
Subskrybuj