Transcriptional analysis of metastatic uveal melanoma survival nominates NRP1 as a therapeutic target

Riyue Bao, Oliver Surriga, Daniel J Olson, Jacob B Allred, Carrie A Strand, Yuanyuan Zha, Timothy Carll, Brian W Labadie, Bruno R Bastos, Marcus Butler, David Hogg, Elgilda Musi, Grazia Ambrosini, Pamela Munster, Gary K Schwartz, Jason J Luke, Riyue Bao, Oliver Surriga, Daniel J Olson, Jacob B Allred, Carrie A Strand, Yuanyuan Zha, Timothy Carll, Brian W Labadie, Bruno R Bastos, Marcus Butler, David Hogg, Elgilda Musi, Grazia Ambrosini, Pamela Munster, Gary K Schwartz, Jason J Luke

Abstract

Uveal melanoma is a rare form of melanoma with particularly poor outcomes in the metastatic setting. In contrast with cutaneous melanoma, uveal melanoma lacks BRAF mutations and demonstrates very low response rates to immune-checkpoint blockade. Our objectives were to study the transcriptomics of metastatic uveal melanoma with the intent of assessing gene pathways and potential molecular characteristics that might be nominated for further exploration as therapeutic targets. We initially analyzed transcriptional data from The Cancer Genome Atlas suggesting PI3K/mTOR and glycolysis as well as IL6 associating with poor survival. From tumor samples collected in a prospective phase II trial (A091201), we performed a transcriptional analysis of human metastatic uveal melanoma observing a novel role for epithelial-mesenchymal transition associating with survival. Specifically, we nominate and describe initial functional validation of neuropillin-1 from uveal melanoma cells as associated with poor survival and as a mediator of proliferation and migration for uveal melanoma in vitro. These results immediately nominate potential next steps in clinical research for patients with metastatic uveal melanoma.

Trial registration: ClinicalTrials.gov NCT01835145 NCT03565445.

Conflict of interest statement

J.J.L. declares Scientific Advisory Board: (no stock) 7 Hills, Spring bank (stock) Actym, Alphamab Oncology, Arch Oncology, Kanaph, Mavu, Onc.AI, Pyxis, Tempest. Consultancy with compensation: Abbvie, Aligos, Array, Bayer, Bristol-Myers Squibb, Checkmate, Cstone, Eisai, EMD Serono, KSQ, Janssen, Merck, Mersana, Novartis, Partner, Pfizer, RefleXion, Regeneron, Ribon, Rubius, Silicon, Tesaro, Werewolf, Xilio, Xencor. Research Support: (all to institution for clinical trials unless noted) AbbVie, Agios (IIT), Array (IIT), Astellas, Bristol-Myers Squibb (IIT & industry), Corvus, EMD Serono, Immatics, Incyte, Kadmon, Macrogenics, Merck, Spring bank, Tizona, Xencor. Travel: Bristol-Myers Squibb, Janssen, Mersana, Pyxis. Patents: (both provisional) Serial #15/612,657 (Cancer Immunotherapy), PCT/US18/36052 (Microbiome Biomarkers for Anti-PD-1/PD-L1 Responsiveness: Diagnostic, Prognostic and Therapeutic Uses Thereof). RB: None declared, Patents: (all provisional) PCT/US15/612657 (Cancer Immunotherapy), PCT/US18/36052 (Microbiome Biomarkers for Anti-PD-1/PD-L1 Responsiveness: Diagnostic, Prognostic and Therapeutic Uses Thereof), PCT/US63/055227 (Methods and Compositions for Treating Autoimmune and Allergic Disorders). For the remaining authors, there are no conflicts of interest.

Copyright © 2020 The Author(s). Published by Wolters Kluwer Health, Inc.

Figures

Fig. 1
Fig. 1
Transcriptional programs associated with overall survival in primary uveal melanoma (pUM) from The Cancer Genome Atlas (TCGA). (a) Kaplan–Meier survival curves of MTORC1 signaling and IL6/JAK/STAT3 signaling gene expression in pUM, split by median expression of each signature (high vs. low). Survival risk table is shown below the Kaplan–Meier plot in each panel. (b) Forest plots showing the hazard ratio and P-values in Cox proportional hazards (PHs) multivariable model of the signaling pathways with demographic and clinical covariates. n = 80 patients in the TCGA primary UM cohort were shown for (a) and (b). Log-rank test was used in (a), and Cox PH multivariable model was used in (b).
Fig. 2
Fig. 2
Epithelial-mesenchymal transition (EMT) signature is significantly associated with overall survival (OS) in patients with metastatic uveal melanoma. (a) Expression of the EMT gene signature in patient survival groups split by 1-year OS; n = 14 in patients with OS ≤ 1 year, n = 5 in patients with OS > 1 year. (b) Kaplan–Meier survival analysis of OS in patients with tumors EMThigh and EMTlow, split by median expression of the EMT gene signature. Nineteen patients were shown in (a) and (b), split by two different metrics [1-year OS in (a), and median EMT expression in (b)]. Survival risk table is shown below the plot. Two-sided Student’s t-test was used in (a). Cox proportional hazards univariable model was used in (b).
Fig. 3
Fig. 3
Differentially expressed genes of metastatic uveal melanoma in patient survival groups split by 1-year overall survival (OS). (a) Expression heatmap of 76 differentially expressed gens (DEGs) comparing tumors from patients who lived less than 1 year to those who lived longer. Genes were filtered by P < 0.005 (unadjusted) and fold change ≥2.0 or ≤–2.0. Samples were clustered on the column with dendrogram shown above the heatmap. Annotation bar labels patient groups with OS ≤ or >1 year. Genes are shown in the boxes to the right of the heatmap, following the same order as the gene dendrogram to the left side of the heatmap. (b) Expression of 22 DEGs upregulated in tumors from patients with OS ≤ 1 year relative to those with OS > 1 year. FC = expression fold of change calculated by comparing patients with OS >1 year to patients with OS ≤1 year. 1 yr = one year. The limma voom regression model with precision weights was used in (a) and (b).
Fig. 4
Fig. 4
Knockdown of neuropillin-1 (NRP1) gene expression in vitro induces G1 arrest and inhibits cell proliferation and invasion. (a) NRP1 mRNA expression in uveal melanoma cell lines Mel290, Mel285, OMM1.3, and OCM3. n = 2 in each of the four cell lines, quantitative PCR was performed in triplicates, experiment was repeated two times. (b) Western blots of Mel290, Mel285, OMM1.3, and OCM3 cells transfected with NRP1 small interfering RNA (siRNA) shows inhibition of NRP1 expression and induction of p27Kip1 expression. (c) Cell cycle analysis of uveal melanoma cells transfected with NRP1 siRNA for 72 h shows a significant increase in G1 population in Mel290 (P < 0.0001) and Mel285 (P = 0.035) cells but not OMM1.3 and OCM3 cells; n = 3 in each of the four cell lines, with two groups each, experiment was repeated three times. (d and e) The siRNA knockdown of NRP1 expression significantly inhibits cell viability (d) after 72 h (Mel290 P = 0.009, Mel285 P = 0.003, OMM1.3 P = 0.18, OCM3 P < 0.001) and cell invasion (e) after 24 h of uveal melanoma cells that highly express NRP1 (Mel290 and Mel285). In (d), n = 3 in each of the four cell lines, with two groups each, experiment was repeated three times. (f) Quantitation of migrated cells shows the selective inhibition of invasion by NRP1 siRNA knockdown in Mel290 (P = 0.046) and Mel285 (P = 0.007) cells but not OMM1.3 (P = 0.40) and OCM3 (P = 0.42) cells. n = 3 in each of the four cell lines, with two groups each, experiment was repeated three times. In (c), (d), and (f), each bar is shown as mean ± S.E.M, with standard error calculated as the SD divided by the square root of the number of samples. Two-sided Student’s t-test was used in (c), (d), and (f); ****P < 0.0001, ***P < 0.001, **P < 0.01, *P < 0.05.

References

    1. Luke JJ, Triozzi PL, McKenna KC, Van Meir EG, Gershenwald JE, Bastian BC, et al. Biology of advanced uveal melanoma and next steps for clinical therapeutics. Pigment Cell Melanoma Res. 2015; 28:135–147
    1. Onken MD, Worley LA, Ehlers JP, Harbour JW. Gene expression profiling in uveal melanoma reveals two molecular classes and predicts metastatic death. Cancer Res. 2004; 64:7205–7209
    1. Luke JJ, Flaherty KT, Ribas A, Long GV. Targeted agents and immunotherapies: optimizing outcomes in melanoma. Nat Rev Clin Oncol. 2017; 14:463–482
    1. Luke JJ, Callahan MK, Postow MA, Romano E, Ramaiya N, Bluth M, et al. Clinical activity of ipilimumab for metastatic uveal melanoma: a retrospective review of the Dana-Farber Cancer Institute, Massachusetts General Hospital, Memorial Sloan-Kettering Cancer Center, and University Hospital of Lausanne experience. Cancer. 2013; 119:3687–3695
    1. Algazi AP, Tsai KK, Shoushtari AN, Munhoz RR, Eroglu Z, Piulats JM, et al. Clinical outcomes in metastatic uveal melanoma treated with PD-1 and PD-L1 antibodies. Cancer. 2016; 122:3344–3353
    1. Carvajal RD, Piperno-Neumann S, Kapiteijn E, Chapman PB, Frank S, Joshua AM, et al. Selumetinib in combination with dacarbazine in patients with metastatic uveal melanoma: a phase III, multicenter, randomized trial (SUMIT). J Clin Oncol. 2018; 36:1232–1239
    1. Van Raamsdonk CD, Griewank KG, Crosby MB, Garrido MC, Vemula S, Wiesner T, et al. Mutations in GNA11 in uveal melanoma. N Engl J Med. 2010; 363:2191–2199
    1. Van Raamsdonk CD, Bezrookove V, Green G, Bauer J, Gaugler L, O’Brien JM, et al. Frequent somatic mutations of GNAQ in uveal melanoma and blue naevi. Nature. 2009; 457:599–602
    1. Yu FX, Luo J, Mo JS, Liu G, Kim YC, Meng Z, et al. Mutant Gq/11 promote uveal melanoma tumorigenesis by activating YAP. Cancer Cell. 2014; 25:822–830
    1. Feng X, Degese MS, Iglesias-Bartolome R, Vaque JP, Molinolo AA, Rodrigues M, et al. Hippo-independent activation of YAP by the GNAQ uveal melanoma oncogene through a trio-regulated rho GTPase signaling circuitry. Cancer Cell. 2014; 25:831–845
    1. Moore AR, Ceraudo E, Sher JJ, Guan Y, Shoushtari AN, Chang MT, et al. Recurrent activating mutations of G-protein-coupled receptor CYSLTR2 in uveal melanoma. Nat Genet. 2016; 48:675–680
    1. Johansson P, Aoude LG, Wadt K, Glasson WJ, Warrier SK, Hewitt AW, et al. Deep sequencing of uveal melanoma identifies a recurrent mutation in PLCB4. Oncotarget. 2016; 7:4624–4631
    1. Robertson AG, Shih J, Yau C, Gibb EA, Oba J, Mungall KL, et al. ; TCGA Research Network. Integrative analysis identifies four molecular and clinical subsets in uveal melanoma. Cancer Cell. 2017; 32:204–220.e15
    1. Mallikarjuna K, Pushparaj V, Biswas J, Krishnakumar S. Expression of epidermal growth factor receptor, ezrin, hepatocyte growth factor, and c-Met in uveal melanoma: an immunohistochemical study. Curr Eye Res. 2007; 32:281–290
    1. Wu X, Zhou J, Rogers AM, Jänne PA, Benedettini E, Loda M, Hodi FS. c-Met, epidermal growth factor receptor, and insulin-like growth factor-1 receptor are important for growth in uveal melanoma and independently contribute to migration and metastatic potential. Melanoma Res. 2012; 22:123–132
    1. Luke JJ, Olson DJ, Allred JB, Strand CA, Bao R, Zha Y, et al. Randomized phase II trial and tumor mutational spectrum analysis from cabozantinib versus chemotherapy in metastatic uveal melanoma (alliance A091201). Clin Cancer Res. 2020; 26:804–811
    1. Piperno-Neumann S, Kapiteijn E, Larkin JMG, Carvajal RD, Luke JJ, Seifert H, et al. Landscape of genetic alterations in patients with metastatic uveal melanoma. J Clin Oncol. 2014; 3215_Suppl9043–9043
    1. Royer-Bertrand B, Torsello M, Rimoldi D, El Zaoui I, Cisarova K, Pescini-Gobert R, et al. Comprehensive genetic landscape of uveal melanoma by whole-genome sequencing. Am J Hum Genet. 2016; 99:1190–1198
    1. Shain AH, Bagger MM, Yu R, Chang D, Liu S, Vemula S, et al. The genetic evolution of metastatic uveal melanoma. Nat Genet. 2019; 51:1123–1130
    1. Johnson DB, Bao R, Ancell KK, Daniels AB, Wallace D, Sosman JA, Luke JJ. Response to anti-PD-1 in uveal melanoma without high-volume liver metastasis. J Natl Compr Canc Netw. 2019; 17:114–117
    1. Sato T, Nathan PD, Hernandez-Aya LF, Sacco JJ, Orloff MM, Truscello J, et al. Intra-patient escalation dosing strategy with IMCgp100 results in mitigation of T-cell based toxicity and preliminary efficacy in advanced uveal melanoma. J Clin Oncol. 2017; 3515_Suppl9531–9531
    1. Chandran SS, Somerville RPT, Yang JC, Sherry RM, Klebanoff CA, Goff SL, et al. Treatment of metastatic uveal melanoma with adoptive transfer of tumour-infiltrating lymphocytes: a single-centre, two-stage, single-arm, phase 2 study. Lancet Oncol. 2017; 18:792–802
    1. Heppt MV, Amaral T, Kähler KC, Heinzerling L, Hassel JC, Meissner M, et al. Combined immune checkpoint blockade for metastatic uveal melanoma: a retrospective, multi-center study. J Immunother Cancer. 2019; 7:299.
    1. Durante MA, Rodriguez DA, Kurtenbach S, Kuznetsov JN, Sanchez MI, Decatur CL, et al. Single-cell analysis reveals new evolutionary complexity in uveal melanoma. Nat Commun. 2020; 11:496.
    1. Karlsson J, Nilsson LM, Mitra S, Alsén S, Shelke GV, Sah VR, et al. Molecular profiling of driver events in metastatic uveal melanoma. Nat Commun. 2020; 11:1894.
    1. Godfrey J, Tumuluru S, Bao R, Leukam M, Venkataraman G, Phillip J, et al. PD-L1 gene alterations identify a subset of diffuse large B-cell lymphoma harboring a T-cell-inflamed phenotype. Blood. 2019; 133:2279–2290
    1. Andrews S. Babraham Institute Project page; FastQC: A quality control application for high throughput sequence data. . [Accessed 13 September 2018]
    1. Bray NL, Pimentel H, Melsted P, Pachter L. Near-optimal probabilistic RNA-seq quantification. Nat Biotechnol. 2016; 34:525–527
    1. Harrow J, Frankish A, Gonzalez JM, Tapanari E, Diekhans M, Kokocinski F, et al. GENCODE: the reference human genome annotation for The ENCODE Project. Genome Res. 2012; 22:1760–1774
    1. Soneson C, Love MI, Robinson MD. Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Res. 2015; 4:1521.
    1. Law CW, Chen Y, Shi W, Smyth GK. voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 2014; 15:R29.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Statist Soc. 1995; B:289–300
    1. Sergushichev A. An algorithm for fast preranked gene set enrichment analysis using cumulative statistic calculation. bioRxiv. 2016
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005; 102:15545–15550
    1. Zuidervaart W, van Nieuwpoort F, Stark M, Dijkman R, Packer L, Borgstein AM, et al. Activation of the MAPK pathway is a common event in uveal melanomas although it rarely occurs through mutation of BRAF or RAS. Br J Cancer. 2005; 92:2032–2038
    1. Luyten GP, Mooy CM, De Jong PT, Hoogeveen AT, Luider TM. A chicken embryo model to study the growth of human uveal melanoma. Biochem Biophys Res Commun. 1993; 192:22–29
    1. Chu W, Song X, Yang X, Ma L, Zhu J, He M, et al. Neuropilin-1 promotes epithelial-to-mesenchymal transition by stimulating nuclear factor-kappa B and is associated with poor prognosis in human oral squamous cell carcinoma. PLoS One. 2014; 9:e101931.
    1. Luo M, Hou L, Li J, Shao S, Huang S, Meng D, et al. VEGF/NRP-1axis promotes progression of breast cancer via enhancement of epithelial-mesenchymal transition and activation of NF-κB and β-catenin. Cancer Lett. 2016; 373:1–11
    1. Matkar PN, Singh KK, Rudenko D, Kim YJ, Kuliszewski MA, Prud’homme GJ, et al. Novel regulatory role of neuropilin-1 in endothelial-to-mesenchymal transition and fibrosis in pancreatic ductal adenocarcinoma. Oncotarget. 2016; 7:69489–69506
    1. Ma L, Zhai B, Zhu H, Li W, Jiang W, Lei L, et al. The miR-141/neuropilin-1 axis is associated with the clinicopathology and contributes to the growth and metastasis of pancreatic cancer. Cancer Cell Int. 2019; 19:248.
    1. Weekes CD, Beeram M, Tolcher AW, Papadopoulos KP, Gore L, Hegde P, et al. A phase I study of the human monoclonal anti-NRP1 antibody MNRP1685A in patients with advanced solid tumors. Invest New Drugs. 2014; 32:653–660
    1. Yoshida A, Shimizu A, Asano H, Kadonosono T, Kondoh SK, Geretti E, et al. VEGF-A/NRP1 stimulates GIPC1 and Syx complex formation to promote RhoA activation and proliferation in skin cancer cells. Biol Open. 2015; 4:1063–1076
    1. Spranger S, Luke JJ, Bao R, Zha Y, Hernandez KM, Li Y, et al. Density of immunogenic antigens does not explain the presence or absence of the T-cell-inflamed tumor microenvironment in melanoma. Proc Natl Acad Sci U S A. 2016; 113:E7759–E7768
    1. Peng W, Chen JQ, Liu C, Malu S, Creasy C, Tetzlaff MT, et al. Loss of PTEN promotes resistance to T cell-mediated immunotherapy. Cancer Discov. 2016; 6:202–216
    1. Luke JJ, Bao R, Sweis RF, Spranger S, Gajewski TF. WNT/β-catenin pathway activation correlates with immune exclusion across human cancers. Clin Cancer Res. 2019; 25:3074–3083
    1. Dongre A, Weinberg RA. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat Rev Mol Cell Biol. 2019; 20:69–84
    1. Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature. 2015; 523:231–235
    1. Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell. 2016; 165:35–44
    1. Soker S, Takashima S, Miao HQ, Neufeld G, Klagsbrun M. Neuropilin-1 is expressed by endothelial and tumor cells as an isoform-specific receptor for vascular endothelial growth factor. Cell. 1998; 92:735–745
    1. Delgoffe GM, Woo SR, Turnis ME, Gravano DM, Guy C, Overacre AE, et al. Stability and function of regulatory T cells is maintained by a neuropilin-1-semaphorin-4a axis. Nature. 2013; 501:252–256
    1. Roy S, Bag AK, Singh RK, Talmadge JE, Batra SK, Datta K. Multifaceted role of neuropilins in the immune system: potential targets for immunotherapy. Front Immunol. 2017; 8:1228.

Source: PubMed

3
Abonneren