Abstract
Immune checkpoint inhibition has shown success in treating metastatic cutaneous melanoma but has limited efficacy against metastatic uveal melanoma, a rare variant arising from the immune privileged eye. To better understand this resistance, we comprehensively profile 100 human uveal melanoma metastases using clinicogenomics, transcriptomics, and tumor infiltrating lymphocyte potency assessment. We find that over half of these metastases harbor tumor infiltrating lymphocytes with potent autologous tumor specificity, despite low mutational burden and resistance to prior immunotherapies. However, we observe strikingly low intratumoral T cell receptor clonality within the tumor microenvironment even after prior immunotherapies. To harness these quiescent tumor infiltrating lymphocytes, we develop a transcriptomic biomarker to enable in vivo identification and ex vivo liberation to counter their growth suppression. Finally, we demonstrate that adoptive transfer of these transcriptomically selected tumor infiltrating lymphocytes can promote tumor immunity in patients with metastatic uveal melanoma when other immunotherapies are incapable.
Data availability
Bulk total RNAseq raw sequencing data generated in this study have been deposited in the database of Genotypes and Phenotypes (dbGaP) under accession number phs003330.v1.p1 [https://www.ncbi.nlm.nih.gov/projects/gap/cgi-bin/study.cgi?study_id=phs003330.v1.p1]. These data are available under restricted access for patient confidentiality reasons and access can be obtained by request via the dbGaP system by following the instructions provided by the website. Approval is determined by the National Cancer Institute Data Access Committee, which can be emailed at ncidac@mail.nih.gov. Access to data is generally granted within a month of successful application and available indefinitely thereafter. Selected raw data are protected and are not publicly available due to data privacy laws but may be shared upon request. Source data are provided with this paper.
Code availability
Software packages were implemented as described in the “Methods” and no custom packages were created.
Acknowledgements
We thank the members of the UPMC clinical trial and research teams for their efforts in this study and the Surgery Branch, NCI, for providing selective tumor samples. We thank Clinigen for providing interleukin-2 for clinical and research studies and thank all the patients who participated in this study. This research was supported by the UPMC Immune Transplant and Therapy Center and in part by the University of Pittsburgh Center for Research Computing, RRID:SCR_022735, through the resources provided. Specifically, this work used the HTC cluster, which is supported by NIH award number S10OD028483. This work utilized the UPMC Hillman Cancer Center Immunologic Monitoring and Cellular Products Laboratory, a shared resource at the University of Pittsburgh supported by the CCSG P30 CA047904. S.L.-M. was supported by a National Institutes of Health National Cancer Institute training grant (T32CA113263).
Author information
Authors and Affiliations
Contributions
S.L.-M., C.B., G.Y., S.M., C.G., J.T., E.A. and P.M. carried out the experiments. S.L.-M., R.K. and U.K. analyzed data and prepared visualizations. S.L.-M., P.M., A.C., V.S., P.C. and U.C. designed the RNAseq bioinformatic pipeline. S.L.-M., P.M., E.S., G.V., L.K., D.K. and X.Z. performed next-generation sequencing. U.K. conducted associated TIL ACT clinical trials. Y.-M.H. and U.K. supervised TIL manufacturing associated with TIL ACT clinical trials. S.L.-M. collected clinicogenomic patient and metastasis data. S.L.-M. and U.K. conceived the project, designed the experiments, and wrote the manuscript.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Ethical approval
We have complied with all relevant ethical regulations in the conduct and reporting of this study.
References
-
Hodi, F. S. et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 363, 711–723 (2010).
-
Larkin, J. et al. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 381, 1535–1546 (2019).
-
Samstein, R. M. et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat. Genet. 51, 202–206 (2019).
-
Yarchoan, M., Hopkins, A. & Jaffee, E. M. Tumor mutational burden and response rate to PD-1 inhibition. N. Engl. J. Med. 377, 2500–2501 (2017).
-
Tawbi, H. A. et al. Relatlimab and nivolumab versus nivolumab in untreated advanced melanoma. N. Engl. J. Med. 386, 24–34 (2022).
-
Kalbasi, A. & Ribas, A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat. Rev. Immunol. 20, 25–39 (2020).
-
Algazi, A. P. et al. Clinical outcomes in metastatic uveal melanoma treated with PD-1 and PD-L1 antibodies. Cancer 122, 3344–3353 (2016).
-
Piulats, J. M. et al. Nivolumab plus ipilimumab for treatment-naïve metastatic uveal melanoma: an open-label, multicenter, phase II trial by the Spanish Multidisciplinary Melanoma Group (GEM-1402). J. Clin. Oncol. 39, 586–598 (2021).
-
Jager, M. J. et al. Uveal melanoma. Nat. Rev. Dis. Primers 6, 24 (2020).
-
Singh, A. D., Turell, M. E. & Topham, A. K. Uveal melanoma: trends in incidence, treatment, and survival. Ophthalmology 118, 1881–1885 (2011).
-
Niederkorn, J. Y. Ocular immune privilege and ocular melanoma: parallel universes or immunological plagiarism? Front. Immunol. 3, 148 (2012).
-
Carvajal, R. D. et al. Clinical and molecular response to tebentafusp in previously treated patients with metastatic uveal melanoma: a phase 2 trial. Nat. Med. 28, 2364–2373 (2022).
-
Nathan, P. et al. Overall survival benefit with tebentafusp in metastatic uveal melanoma. N. Engl. J. Med. 385, 1196–1206 (2021).
-
Rothermel, L. D. et al. Identification of an immunogenic subset of metastatic uveal melanoma. Clin. Cancer Res. 22, 2237–2249 (2016).
-
Chandran, S. S. 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. 18, 792–802 (2017).
-
Crompton, J. G., Klemen, N. & Kammula, U. S. Metastasectomy for tumor-infiltrating lymphocytes: an emerging operative indication in surgical oncology. Ann. Surg. Oncol. 25, 565–572 (2018).
-
Javed, A. et al. PD-L1 expression in tumor metastasis is different between uveal melanoma and cutaneous melanoma. Immunotherapy 9, 1323–1330 (2017).
-
Foroutan, M. et al. Single sample scoring of molecular phenotypes. BMC Bioinform. 19, 404 (2018).
-
Nieto, P. et al. A single-cell tumor immune atlas for precision oncology. Genome Res. 31, 1913–1926 (2021).
-
Andreatta, M. & Carmona, S. J. UCell: robust and scalable single-cell gene signature scoring. Comput. Struct. Biotechnol. J. 19, 3796–3798 (2021).
-
Kang, J. B. et al. Efficient and precise single-cell reference atlas mapping with Symphony. Nat. Commun. 12, 5890 (2021).
-
Smith-Garvin, J. E., Koretzky, G. A. & Jordan, M. S. T cell activation. Annu. Rev. Immunol. 27, 591–619 (2009).
-
Ivashkiv, L. B. IFNγ: signalling, epigenetics and roles in immunity, metabolism, disease and cancer immunotherapy. Nat. Rev. Immunol. 18, 545–558 (2018).
-
Dersh, D., Hollý, J. & Yewdell, J. W. A few good peptides: MHC class I-based cancer immunosurveillance and immunoevasion. Nat. Rev. Immunol. 21, 116–128 (2021).
-
Diamond, M. S. & Farzan, M. The broad-spectrum antiviral functions of IFIT and IFITM proteins. Nat. Rev. Immunol. 13, 46–57 (2013).
-
Chandran, S. S. et al. Tumor-specific effector CD8+ T cells that can establish immunological memory in humans after adoptive transfer are marked by expression of IL7 receptor and c-myc. Cancer Res. 75, 3216–3226 (2015).
-
Krishna, S. et al. Stem-like CD8 T cells mediate response of adoptive cell immunotherapy against human cancer. Science 370, 1328–1334 (2020).
-
Chow, M. T. et al. Intratumoral activity of the CXCR3 chemokine system is required for the efficacy of anti-PD-1 therapy. Immunity 50, 1498–1512.e5 (2019).
-
Chen, J. et al. SLAMF7 is critical for phagocytosis of haematopoietic tumour cells via Mac-1 integrin. Nature 544, 493–497 (2017).
-
Spranger, S., Bao, R. & Gajewski, T. F. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 523, 231–235 (2015).
-
Duraiswamy, J. et al. Myeloid antigen-presenting cell niches sustain antitumor T cells and license PD-1 blockade via CD28 costimulation. Cancer Cell 39, 1623–1642.e20 (2021).
-
Luke, J. J., Bao, R., Sweis, R. F., Spranger, S. & Gajewski, T. F. WNT/β-catenin pathway activation correlates with immune exclusion across human cancers. Clin Cancer Res. 25, 3074–3083 (2019).
-
Jerby-Arnon, L. et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell 175, 984–997.e24 (2018).
-
Yu, F. et al. Loss of lncRNA-SNHG7 promotes the suppression of hepatic stellate cell activation via miR-378a-3p and DVL2. Mol. Ther. Nucleic Acids 17, 235–244 (2019).
-
Chen, Y. et al. Knockdown of lncRNA SNHG7 inhibited cell proliferation and migration in bladder cancer through activating Wnt/β-catenin pathway. Pathol. Res. Pract. 215, 302–307 (2019).
-
Bian, Z. et al. The role of long noncoding RNA SNHG7 in human cancers (Review). Mol. Clin. Oncol. 13, 45 (2020).
-
Najafi, S. et al. Oncogenic roles of small nucleolar RNA host gene 7 (SNHG7) long noncoding RNA in human cancers and potentials. Front. Cell Dev. Biol. 9, 809345 (2021).
-
Ren, J. et al. Long noncoding RNA SNHG7 promotes the progression and growth of glioblastoma via inhibition of miR-5095. Biochem. Biophys. Res. Commun. 496, 712–718 (2018).
-
Rooney, M. S., Shukla, S. A., Wu, C. J., Getz, G. & Hacohen, N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell 160, 48–61 (2015).
-
Fehrenbacher, L. et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet 387, 1837–1846 (2016).
-
Ayers, M. et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Invest. 127, 2930–2940 (2017).
-
Chiffelle, J. et al. T-cell repertoire analysis and metrics of diversity and clonality. Curr. Opin. Biotechnol. 65, 284–295 (2020).
-
Robertson, A. G. et al. Integrative analysis identifies four molecular and clinical subsets in uveal melanoma. Cancer Cell 32, 204–220.e15 (2017).
-
Field, M. G. et al. Punctuated evolution of canonical genomic aberrations in uveal melanoma. Nat. Commun. 9, 116 (2018).
-
Carvajal, R. D. et al. Advances in the clinical management of uveal melanoma. Nat. Rev. Clin. Oncol. 20, 99–115 (2023).
-
Pelster, M. S. et al. Nivolumab and ipilimumab in metastatic uveal melanoma: results from a single-arm phase II study. J. Clin. Oncol. 39, 599–607 (2021).
-
Karlsson, J. et al. Molecular profiling of driver events in metastatic uveal melanoma. Nat. Commun. 11, 1894 (2020).
-
Shain, A. H. et al. The genetic evolution of metastatic uveal melanoma. Nat. Genet. 51, 1123–1130 (2019).
-
Durante, M. A. et al. Single-cell analysis reveals new evolutionary complexity in uveal melanoma. Nat. Commun. 11, 496 (2020).
-
Spranger, S., Dai, D., Horton, B. & Gajewski, T. F. Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell 31, 711–723.e14 (2017).
-
Sharma, P., Hu-Lieskovan, S., Wargo, J. A. & Ribas, A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 168, 707–723 (2017).
-
Wang, Y. et al. Multimodal single-cell and whole-genome sequencing of small, frozen clinical specimens. Nat. Genet. 55, 19–25 (2023).
-
Bugter, J. M., Fenderico, N. & Maurice, M. M. Mutations and mechanisms of WNT pathway tumour suppressors in cancer. Nat. Rev. Cancer 21, 5–21 (2021).
-
Mariani, P. et al. Immunohistochemical characterisation of the immune landscape in primary uveal melanoma and liver metastases. Br. J. Cancer 129, 772–781 (2023).
-
Valpione, S. et al. The T cell receptor repertoire of tumor infiltrating T cells is predictive and prognostic for cancer survival. Nat. Commun. 12, 4098 (2021).
-
Yusko, E. et al. Association of tumor microenvironment T-cell repertoire and mutational load with clinical outcome after sequential checkpoint blockade in melanoma. Cancer Immunol. Res. 7, 458–465 (2019).
-
Riaz, N. et al. Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell 171, 934–949.e16 (2017).
-
Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 568–571 (2014).
-
Hanada, K. I. et al. A phenotypic signature that identifies neoantigen-reactive T cells in fresh human lung cancers. Cancer Cell 40, 479–493.e6 (2022).
-
Lowery, F. J. et al. Molecular signatures of antitumor neoantigen-reactive T cells from metastatic human cancers. Science 375, 877–884 (2022).
-
Eisenhauer, E. A. et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur. J. Cancer 45, 228–247 (2009).
-
Kendig, K. I. et al. Sentieon DNASeq variant calling workflow demonstrates strong computational performance and accuracy. Front. Genet. 10, 736 (2019).
-
Van der Auwera, G. A. et al. From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr. Protoc. Bioinform. 43, 11.10.11–11.10.33 (2013).
-
Talevich, E., Shain, A. H., Botton, T. & Bastian, B. C. CNVkit: genome-wide copy number detection and visualization from targeted DNA sequencing. PLoS Comput. Biol. 12, e1004873 (2016).
-
Abyzov, A., Urban, A. E., Snyder, M. & Gerstein, M. CNVnator: an approach to discover, genotype, and characterize typical and atypical CNVs from family and population genome sequencing. Genome Res. 21, 974–984 (2011).
-
Chen, X. et al. Manta: rapid detection of structural variants and indels for germline and cancer sequencing applications. Bioinformatics 32, 1220–1222 (2016).
-
Jeffares, D. C. et al. Transient structural variations have strong effects on quantitative traits and reproductive isolation in fission yeast. Nat. Commun. 8, 14061 (2017).
-
Geoffroy, V. et al. AnnotSV: an integrated tool for structural variations annotation. Bioinformatics 34, 3572–3574 (2018).
-
Kandoth, C. mskcc/vcf2maf: vcf2maf v1.6.19 (2020).
-
McLaren, W. et al. The Ensembl variant effect predictor. Genome Biol. 17, 122 (2016).
-
Smigielski, E. M., Sirotkin, K., Ward, M. & Sherry, S. T. dbSNP: a database of single nucleotide polymorphisms. Nucleic Acids Res. 28, 352–355 (2000).
-
Tate, J. G. et al. COSMIC: the Catalogue Of Somatic Mutations In Cancer. Nucleic Acids Res. 47, D941–D947 (2019).
-
Mayakonda, A., Lin, D. C., Assenov, Y., Plass, C. & Koeffler, H. P. Maftools: efficient and comprehensive analysis of somatic variants in cancer. Genome Res. 28, 1747–1756 (2018).
-
Paniccia, A. et al. Prospective, multi-institutional, real-time next-generation sequencing of pancreatic cyst fluid reveals diverse genomic alterations that improve the clinical management of pancreatic cysts. Gastroenterology 164, 117–133.e17 (2023).
-
Andrews, S. FastQC: a quality control tool for high throughput sequence data (2010).
-
Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 17, 10–12 (2011).
-
Kopylova, E., Noé, L. & Touzet, H. SortMeRNA: fast and accurate filtering of ribosomal RNAs in metatranscriptomic data. Bioinformatics 28, 3211–3217 (2012).
-
Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).
-
Frankish, A. et al. GENCODE 2021. Nucleic Acids Res. 49, D916–D923 (2021).
-
Li, H. et al. The sequence alignment/map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).
-
Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).
-
Cunningham, F. et al. Ensembl 2022. Nucleic Acids Res. 50, D988–D995 (2022).
-
Smedley, D. et al. The BioMart community portal: an innovative alternative to large, centralized data repositories. Nucleic Acids Res. 43, W589–598 (2015).
-
Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).
-
Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).
-
Blighe, K. PCAtools: PCAtools: Everything Principal Components Analysis. R package version 2.10.0 (2022).
-
Hänzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinform. 14, 7 (2013).
-
Sherman, B. T. et al. DAVID: a web server for functional enrichment analysis and functional annotation of gene lists (2021 update). Nucleic Acids Res. 50, W216–221 (2022).
-
Wu, T. et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation 2, 100141 (2021).
-
Liberzon, A. et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 1, 417–425 (2015).
-
The Gene Ontology Consortium. The Gene Ontology Resource: 20 years and still GOing strong. Nucleic Acids Res. 47, D330–D338 (2019).
-
Orenbuch, R. et al. arcasHLA: high-resolution HLA typing from RNAseq. Bioinformatics 36, 33–40 (2020).
-
Bolotin, D. A. et al. MiXCR: software for comprehensive adaptive immunity profiling. Nat. Methods 12, 380–381 (2015).
-
Nazarov, V. et al. immunarch: Bioinformatics Analysis of T-Cell and B-Cell Immune Repertoires R package version 1.0.0 (2022).
-
Zheng, G. X. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).
-
Young, M. D. & Behjati, S. SoupX removes ambient RNA contamination from droplet-based single-cell RNA sequencing data. Gigascience 9, 12 (2020)
-
Choudhary, S. & Satija, R. Comparison and evaluation of statistical error models for scRNA-seq. Genome Biol. 23, 27 (2022).
-
Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e29 (2021).
-
McGinnis, C. S., Murrow, L. M. & Gartner, Z. J. DoubletFinder: doublet detection in single-cell RNA sequencing data using artificial nearest neighbors. Cell Syst. 8, 329–337.e4 (2019).
-
Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with harmony. Nat. Methods 16, 1289–1296 (2019).
-
Phipson, B. et al. propeller: testing for differences in cell type proportions in single cell data. Bioinformatics 38, 4720–4726 (2022).
-
Borcherding, N., Bormann, N. L. & Kraus, G. scRepertoire: an R-based toolkit for single-cell immune receptor analysis. F1000Res 9, 47 (2020).
-
The R Development Core Team. R: A Language and Environment for Statistical Computing (2021).
-
RStudio Team. RStudio: Integrated Development for R (2020).
-
Kassambara, A. et al. survminer: Drawing Survival Curves using ‘ggplot2’. R package version 0.4.9 (2021).
-
Gu, Z., Eils, R. & Schlesner, M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 32, 2847–2849 (2016).
-
Wickham, H. et al. Welcome to the tidyverse. J. Open Source Softw. 4, 1686 (2019).
-
Wickham, H. ggplot2: Elegant Graphics for Data Analysis (Springer-Verlag, 2016).
-
Brewer, C. RColorBrewer: ColorBrewer Palettes. R package version 1.1-3 (2022).
-
Dawson, C. ggprism: A. ‘ggplot2’ Extension Inspired by ‘GraphPad Prism’. R package version 1.0.4 (2022).
-
Pedersen, T. patchwork: The Composer of Plots. R package version 1.1.2 (2022).
-
Bedward, M., Eppstein, D. & Menzel, P. packcircles: Circle Packing. R package version 0.3.5 (2022).
Plaats een reactie ...
Reageer op "Uitgezaaide oogmelanoom bevat soms immuuncelkenmerken die patienten gevoelig kan maken voor TIL therapie (adoptieve tumor-infiltrerende lymfocyten - TIL)"