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.

  • Letter
  • Published:

Evolution of delayed resistance to immunotherapy in a melanoma responder

Abstract

Despite initial responses1,2,3, most melanoma patients develop resistance4 to immune checkpoint blockade (ICB). To understand the evolution of resistance, we studied 37 tumor samples over 9 years from a patient with metastatic melanoma with complete clinical response to ICB followed by delayed recurrence and death. Phylogenetic analysis revealed co-evolution of seven lineages with multiple convergent, but independent resistance-associated alterations. All recurrent tumors emerged from a lineage characterized by loss of chromosome 15q, with post-treatment clones acquiring additional genomic driver events. Deconvolution of bulk RNA sequencing and highly multiplexed immunofluorescence (t-CyCIF) revealed differences in immune composition among different lineages. Imaging revealed a vasculogenic mimicry phenotype in NGFRhi tumor cells with high PD-L1 expression in close proximity to immune cells. Rapid autopsy demonstrated two distinct NGFR spatial patterns with high polarity and proximity to immune cells in subcutaneous tumors versus a diffuse spatial pattern in lung tumors, suggesting different roles of this neural-crest-like program in different tumor microenvironments. Broadly, this study establishes a high-resolution map of the evolutionary dynamics of resistance to ICB, characterizes a de-differentiated neural-crest tumor population in melanoma immunotherapy resistance and describes site-specific differences in tumor–immune interactions via longitudinal analysis of a patient with melanoma with an unusual clinical course.

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: Integrated clinical course and phylogenetic characterization of longitudinal tumor biopsies.
Fig. 2: Analysis of immune microenvironment by lineage and time.
Fig. 3: Spatial and immune correlates of NGFRhi tumor cells.
Fig. 4: NGFRhi tumor and immune microenvironment by metastatic sites.

Similar content being viewed by others

Data availability

All requests for raw and analyzed data and materials will be promptly reviewed by the senior author (G.M.B.) to verify whether the request is subject to any intellectual property or confidentiality obligations. Patient-related data not included in the paper may be subject to patient confidentiality. Any data and materials that can be shared will be released via a material transfer agreement. All analyzed sequencing data are in supplementary data available at the journal website and any additional data made publicly available after publication will be found at https://github.com/davidliu-lab/Pt98. All t-CyCIF tissue images are available for online viewing: https://www.cycif.org/data/liu-lin-2019/. Raw sequencing data have been deposited into dbGAP, phs001427.v2.p1, which is publicly accessible. Matched clinical and sequencing characteristics of tumors are in Supplementary Table 1.

References

  1. Wolchok, J. D. et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 377, 1345–1356 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Robert, C. et al. Pembrolizumab versus ipilimumab in advanced melanoma. N. Engl. J. Med. 372, 2521–2532 (2015).

    Article  CAS  PubMed  Google Scholar 

  3. Larkin, J. et al. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 381, 1535–1546 (2019).

    Article  CAS  PubMed  Google Scholar 

  4. Sharma, P., Hu-Lieskovan, S., Wargo, J. A. & Ribas, A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 168, 707–723 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Spranger, S., Bao, R. & Gajewski, T. F. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 523, 231 (2015).

    Article  CAS  PubMed  Google Scholar 

  6. George, S. et al. Loss of PTEN is associated with resistance to anti-PD-1 checkpoint blockade therapy in metastatic uterine leiomyosarcoma. Immunity 46, 197–204 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Xiao, Q. et al. DKK2 imparts tumor immunity evasion through β-catenin-independent suppression of cytotoxic immune-cell activation. Nat. Med. 24, 262–270 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Peng, W. et al. Loss of PTEN promotes resistance to T cell-mediated immunotherapy. Cancer Discov. 6, 202–216 (2016).

    Article  CAS  PubMed  Google Scholar 

  9. Zaretsky, J. M. et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N. Engl. J. Med. 375, 819–829 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Sade-Feldman, M. et al. Resistance to checkpoint blockade therapy through inactivation of antigen presentation. Nat Commun. 8, 1136 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Gao, J. et al. Loss of IFN-γ pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy. Cell 167, 397–404 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Davoli, T., Uno, H., Wooten, E. C. & Elledge S. J. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science https://doi.org/10.1126/science.aaf8399 (2017).

  13. Roh, W. et al. Integrated molecular analysis of tumor biopsies on sequential CTLA-4 and PD-1 blockade reveals markers of response and resistance. Sci. Transl. Med. https://doi.org/10.1126/scitranslmed.aah3560 (2017).

  14. Goel, S. et al. CDK4/6 inhibition triggers anti-tumour immunity. Nature 548, 471 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Rambow, F. et al. Toward minimal residual disease-directed therapy in melanoma. Cell 174, 843–855 (2018).

    Article  CAS  PubMed  Google Scholar 

  16. Restivo, G. et al. The low affinity neurotrophin receptor CD271 regulates phenotype switching in melanoma. Nat. Commun. 8, 1988 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Jerby-Arnon, L. et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell 175, 984–997 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Burrell, R. A., McGranahan, N., Bartek, J. & Swanton, C. The causes and consequences of genetic heterogeneity in cancer evolution. Nature 501, 338–345 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. Gerlinger, M. et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366, 883–892 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Jamal-Hanjani, M. et al. Tracking the evolution of non–small-cell lung cancer. N. Engl. J. Med. 376, 2109–2121 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Faltas, B. M. et al. Clonal evolution of chemotherapy-resistant urothelial carcinoma. Nat. Genet. https://doi.org/10.1038/ng.3692 (2016).

  22. Andor, N. et al. Pan-cancer analysis of the extent and consequences of intratumor heterogeneity. Nat. Med. 22, 105–113 (2016).

    Article  CAS  PubMed  Google Scholar 

  23. Ding, L. et al. Clonal architectures and driver mutations in metastatic melanomas. PLoS ONE 9, e111153 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Jiménez-Sánchez, A. et al. Heterogeneous tumor-immune microenvironments among differentially growing metastases in an ovarian cancer patient. Cell 170, 927–938 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Liu, D. et al. Mutational patterns in chemotherapy resistant muscle-invasive bladder cancer. Nat. Commun. 8, 2193 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Juric, D. et al. Convergent loss of PTEN leads to clinical resistance to a PI(3)Kα inhibitor. Nature 518, 240–244 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Chen, P.-L. et al. Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade. Cancer Discov. 6, 827–837 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Edwards, J. et al. Prevalence and cellular distribution of novel immune checkpoint targets across longitudinal specimens in treatment-naive melanoma patients: implications for clinical trials. Clin. Cancer Res. 25, 3247–3258 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Dewhurst, S. M. et al. Tolerance of whole-genome doubling propagates chromosomal instability and accelerates cancer genome evolution. Cancer Discov. 4, 175–185 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. The Cancer Genome Atlas. Genomic classification of cutaneous melanoma. Cell 161, 1681–1696 (2015).

    Article  Google Scholar 

  31. Liu, D. et al. Integrative molecular and clinical modeling of clinical outcomes to PD1 blockade in patients with metastatic melanoma. Nat. Med. 25, 1916–1927 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Barbie, D. A. et al. Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature 462, 108–112 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Liberzon, A. et al. The molecular signatures database hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Maniotis, A. J. et al. Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am. J. Pathol. 155, 739–752 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Landsberg, J. et al. Melanomas resist T-cell therapy through inflammation-induced reversible dedifferentiation. Nature 490, 412–416 (2012).

    Article  CAS  PubMed  Google Scholar 

  36. Sottoriva, A. et al. A Big Bang model of human colorectal tumor growth. Nat. Genet. 47, 209–216 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Riaz, N. et al. Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell https://doi.org/10.1016/j.cell.2017.09.028 (2017).

  39. Lehraiki, A. et al. Increased CD271 expression by the NF-κB pathway promotes melanoma cell survival and drives acquired resistance to BRAF inhibitor vemurafenib. Cell Discov. 1, 1–13 (2015).

    Article  Google Scholar 

  40. Shaffer, S. M. et al. Rare cell variability and drug-induced reprogramming as a mode of cancer drug resistance. Nature 546, 431–435 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Boshuizen, J. et al. Reversal of pre-existing NGFR-driven tumor and immune therapy resistance. Nat. Commun. 11, 3946 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Valyi-Nagy, K., Kormos, B., Ali, M., Shukla, D. & Valyi-Nagy, T. Stem cell marker CD271 is expressed by vasculogenic mimicry-forming uveal melanoma cells in three-dimensional cultures. Mol. Vis. 18, 588–592 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Tan, L. Y. et al. Control of immune cell entry through the tumour vasculature: a missing link in optimising melanoma immunotherapy? Clin. Transl. Immunol. 6, e134 (2017).

    Article  Google Scholar 

  44. Shi, A. et al. Plasma-derived extracellular vesicle analysis and deconvolution enable prediction and tracking of melanoma checkpoint blockade outcome. Sci. Adv. 6, eabb3461 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Marsavela, G. et al. Circulating tumor DNA predicts outcome from first-, but not second-line treatment and identifies melanoma patients who may benefit from combination immunotherapy. Clin. Cancer Res. 26, 5926–5933 (2020).

    Article  CAS  PubMed  Google Scholar 

  46. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Van Allen, E. M. et al. Whole-exome sequencing and clinical interpretation of formalin-fixed, paraffin-embedded tumor samples to guide precision cancer medicine. Nat. Med. 20, 682–688 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Fisher, S. et al. A scalable, fully automated process for construction of sequence-ready human exome targeted capture libraries. Genome Biol. 12, R1 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Gnirke, A. et al. Solution hybrid selection with ultra-long oligonucleotides for massively parallel targeted sequencing. Nat. Biotechnol. 27, 182–189 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Cibulskis, K. et al. ContEst: estimating cross-contamination of human samples in next generation sequencing data. Bioinformatics 27, 2601–2602 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Lawrence, M. S. et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 505, 495–501 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Hodis, E. et al. A landscape of driver mutations in melanoma. Cell 150, 251–263 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Cibulskis, K. et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 31, 213–219 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Saunders, C. T. et al. Strelka: accurate somatic small-variant calling from sequenced tumor–normal sample pairs. Bioinformatics 28, 1811–1817 (2012).

    Article  CAS  PubMed  Google Scholar 

  55. Ramos, A. H. et al. Oncotator: cancer variant annotation tool. Hum. Mutat. 36, E2423–E2429 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Costello, M. et al. Discovery and characterization of artifactual mutations in deep coverage targeted capture sequencing data due to oxidative DNA damage during sample preparation. Nucleic Acids Res. 41, e67 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Olshen, A. B., Venkatraman, E. S., Lucito, R. & Wigler, M. Circular binary segmentation for the analysis of array-based DNA copy number data. Biostatistics 5, 557–572 (2004).

    Article  PubMed  Google Scholar 

  58. Shen, R., Olshen, A. B. & Ladanyi, M. Integrative clustering of multiple genomic data types using a joint latent variable model with application to breast and lung cancer subtype analysis. Bioinformatics 25, 2906–2912 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Carter, S. L. et al. Absolute quantification of somatic DNA alterations in human cancer. Nat. Biotechnol. 30, 413–421 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Brastianos, P. K. et al. Genomic characterization of brain metastases reveals branched evolution and potential therapeutic targets. Cancer Discov. 5, 1164–1177 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Burrell, R. A. et al. Replication stress links structural and numerical cancer chromosomal instability. Nature 494, 492–496 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. 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 

  63. Leshchiner, I. et al. Comprehensive analysis of tumour initiation, spatial and temporal progression under multiple lines of treatment. Preprint at bioRxiv https://doi.org/10.1101/508127 (2018).

  64. Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-seq data with or without a reference genome. BMC Bioinf. 12, 323 (2011).

    Article  CAS  Google Scholar 

  65. Shukla, S. A. et al. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat. Biotechnol. 33, 1152 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Jurtz, V. et al. NetMHCpan-4.0: improved peptide-MHC class I interaction predictions integrating eluted ligand and peptide binding affinity data. J. Immunol. 199, 3360–3368 (2017).

    Article  CAS  PubMed  Google Scholar 

  67. Hundal, J. et al. pVAC-seq: a genome-guided in silico approach to identifying tumor neoantigens. Genome Med. 8, 11 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Lin, J.-R. et al. Highly multiplexed immunofluorescence imaging of human tissues and tumors using t-CyCIF and conventional optical microscopes. eLife https://doi.org/10.7554/eLife.31657 (2018).

  69. Lin, J.-R., Fallahi-Sichani, M. & Sorger, P. K. Highly multiplexed imaging of single cells using a high-throughput cyclic immunofluorescence method. Nat. Commun. 6, 8390 (2015).

    Article  CAS  PubMed  Google Scholar 

  70. Amir, E. D. et al. viSNE enables visualization of high dimensional single-cell data and reveals phenotypic heterogeneity of leukemia. Nat. Biotechnol. 31, 545–552 (2013).

    Article  CAS  PubMed Central  Google Scholar 

  71. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  73. DeTomaso, D. et al. Functional interpretation of single cell similarity maps. Nat. Commun. 10, 4376 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the Dr. Miriam and Sheldon G. Adelson Medical Research Foundation (M.H.), the Harvard Ludwig Center for Cancer Research, the National Science Foundation (grant no. DGE-1144152, Graduate Research Fellowship no. 2016226995), the Conquer Cancer Foundation Young Investigator Award (D.L.), the Damon Runyon Cancer Research Foundation (Physician Scientist Training Grant (D.L.), Postdoctoral Fellowship (DRQ-03-20, D.S.)), the National Institutes of Health (grants R01CA227388, K08CA222663 (B.I.), K08CA234458 (D.L.) and U54-CA225088 (B.I., J.L., S.W., S.M., D.S. and P.K.S.), P01 CA114046 (M.H.), P50 CA174523 (M.H.), U54 CA224070 (M.H.)), DoD – PRCRP W81XWH-16-1-0119 (CA150619) (M.H.), DoD–CDMRP W81XWH-19-1-0143 (G.M.B.), BroadNext10 (E.M.V.A.), Society for Immunotherapy of Cancer Translational Postdoctoral Fellowship (D.L.), the Burroughs Wellcome Fund Career Award for Medical Scientists (B.I.), the Louis V. Gerstner, Jr Scholars Program (B.I.), the Velocity Fellow Program (B.I.), Abeloff V foundation scholar grant (B.I.), the Barr Award for Innovative Translational Research, Early Postdoc Mobility fellowship (no. P2ZHP3_181475) (D.S.) and the Swiss National Science Foundation (no. P2ZHP3_178022) (M.M).

Author information

Authors and Affiliations

Authors

Contributions

D. Liu, J.R.L., M.K., P.K.S. and G.M.B. conceived and designed the overall study. D.T.F., G.G.K., R.F., T.M., D. Lawrence, D.P.C., T.K. and T.S. collected and reviewed all clinical data. D.T.F. and T.S. performed sample processing and shipping. D.P.C., P.K.B., C.A. and J.H.S. provided samples and clinical annotation. A.T. oversaw sample processing and sequencing. D. Liu and E.R. designed and performed mutational and copy-number analyses. D. Liu, A.S., A.S., A.K., K.N., G.Z. and S.M. designed and performed RNA-seq analysis, including quality control, normalization, signature analysis, CIBERSORT and ssGSEA. D. Liu, E.R., A.H. and M.A.N. performed phylogenetic analyses. M.P.H. and C.L. performed IHC and provided pathologic interpretation. L.L.H. performed single-cell RNA-seq sample processing and E.R. and K.B. analyzed single-cell RNA-seq data. J.R.L., Y.A.C. and M.M. performed t-CyCIF normalization, clustering and interpretation. D.S. and S.W. performed neighborhood analysis. D. Liu, J.R.L., E.R., B.I., M.H., M.A.N., E.M.V.A., K.N., K.T.F., R.J.S., M.K., P.K.S. and G.M.B. oversaw all analyses. D. Liu, J.R.L., P.K.S. and G.M.B. wrote the manuscript and all authors reviewed and approved the final manuscript.

Corresponding author

Correspondence to Genevieve M. Boland.

Ethics declarations

Competing interests

D.L. reports funding by a postdoctoral fellowship from the Society for Immunotherapy of Cancers which is funded in part by an educational grant from Bristol-Meyers Squibb (BMS). BMS has had no input into the conception, conduct or reporting of the submitted work. D.C. has received consulting (GSK, Lilly, Boston Pharmaceuticals) and travel/speaking (Merck) support outside the scope of the present work. G.M.B. had sponsored research agreements with Takeda Oncology, Palleon Pharmaceuticals and Olink Proteomics, which were not used to support this work. She served as a speaker for Novartis and on scientific advisory boards for Nektar Therapeutics and Novartis and consults for Merck, all of which are outside the scope of this work. P.K.B has consulted for Angiochem, Genentech-Roche, Lilly, Tesaro, Voyager Therapeutics, ElevateBio, Pfizer (Array), Pfizer, SK Life Sciences and Dantari, received grant/research support (to Massachusetts General Hospital) from Merck, BMS and Lilly and honoraria from Merck, Pfizer, Genentech-Roche and Lilly, outside the scope of this present work. R.J.S. has served as consultant and/or on Scientific Advisory Boards for Asana Biosciences, AstraZeneca, BMS, Eisai, Iovance, Merck, Novartis, Oncosec, Pfizer, Replimune and reports research funding from Merck outside the scope of this present work. A.S. has received consulting (Lead Pharma, Checkmate and C-reveal Therapeutics) support outside the scope of the present work. K.T.F. has/had served on the Board of Directors of Loxo Oncology, Clovis Oncology, Strata Oncology, Vivid Biosciences, Checkmate Pharmaceuticals and Kinnate Pharmaceuticals; Corporate Advisory Board of X4 Pharmaceuticals; Scientific Advisory Boards of PIC Therapeutics, Sanofi, Amgen, Asana, Adaptimmune, Aeglea, Shattuck Labs, Tolero, Apricity, Oncoceutics, Fog Pharma, Neon, Tvardi, xCures, Monopteros and Vibliome; and as consultant to Lilly, Novartis, Genentech, BMS, Merck, Takeda, Verastem, Boston Biomedical, Pierre Fabre and Debiopharm; and received research funding from Novartis and Sanofi. B.I. is a consultant for Merck and Volastra Therapeutics. A.S. has received consulting (Lead Pharma, Checkmate and C-reveal Therapeutics) support outside the scope of the present work. P.K.S. is a member of the Board of Directors of Applied Biomath and Glencoe Software and member of the SAB for RareCyte and NanoString; he has equity in the first three of these companies. In the last 5 years the Sorger laboratory has received research funding from Novartis and Merck. E.M.V.A. has consulted or advised for Tango Therapeutics, Genome Medical, Invitae, Enara Bio, Janssen, Manifold Bio and Monte Rosa and holds equity in Tango Therapeutics, Genome Medical, Syapse, Enara Bio, Manifold Bio, Microsoft and Monte Rosa; has received research support from Novartis and BMS, travel reimbursement from Roche/Genentech and has Institutional patents filed on chromatin mutations and immunotherapy response and methods for clinical interpretation. All other authors declare no competing interests.

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 Mutation Load and Mutations in Significant Melanoma Genes.

221 nonsynonymous clonal mutations were found in all tumors, including hotspot mutations in IDH1 (p.R132C) and MAP2K1 (p.E203K), and additional missense mutations in cancer driver genes CTNNB1 (p.R582W) and ARID2 (p.P1664S).

Extended Data Fig. 2 Mutational Spectrum Profile of Common Ancestor.

489 common single nucleotide variants (including non-coding mutations) found in all tumors with mutations called individually in each sample are represented in their tri-nucleotide context; this mutational spectrum has cosine similarity of 0.965 with the ultraviolet DNA damage signature (Signature 7; https://cancer.sanger.ac.uk/cosmic/signatures). A similar analysis with 548 common ancestor mutations inferred jointly by PyClone generates similar results with cosine similarity of 0.962 to the UV signature (not shown).

Extended Data Fig. 3 Copy Number Alterations by Lineage and Tumor.

Each bar represents a tumor, with numbers indicating chromosomes and copy number alterations indicated by shade. Red arrows indicate the chromosomal segment loss of heterozygosity with corresponding loss of mutations in that segment that characterize the lineage, that is 2q for Lineage 2, 15q in Lineage 3, 19q in Lineage 5. Genome doubling is indicated by *.

Extended Data Fig. 4 Inferred Lineage-Defining Mutational Clusters and Cancer Cell Fractions by Tumor.

a, Inferred Mutational Clusters and Cluster Cancer Cell Fraction by Tumor. Mutational clusters representing subclones and the proportion of cancer cells in each tumor sample with each mutational cluster was inferred using PyClone. The x-axis shows each tumor, the y-axis is the proportion of cancer cells in each sample containing the cluster, and the legend (n=xx) refers to the number of mutations for each inferred cluster. Only clusters containing more than 3 mutations were included (9 clusters excluded) in subsequent analyses. b, Mutational Cluster Defining Lineage 3. This pattern demonstrates the loss of mutations in a common ancestor of the seventeen tumors in Lineage 3 (T11, T21, the early escape lesions (R1 and R2) and late-emerging resistant lesions (R2 and R3.2) and the post-autopsy lesions). These mutations are all found in chromosome 15, with a corresponding LOH in chromosome 15q. c, Mutational Cluster Defining Early Resistant Small Bowel Metastasis. This cluster represents the acquired mutations shared between the small bowel metastasis (R1) and the other late resistant tumors which also share a bi-allelic CDKN2A deletion. 2/4 mutations were inferred to have multiplicity of 2, and 2/4 multiplicity of 1, consistent with a unique genome doubling event just prior to the emergence of this tumor in lineage 3, and present in all subsequent resistant tumors. These mutations were manually reviewed and showed no evidence of artifact, although MYO7A is detectable at a lower level in P4 than the subsequent resistant tumors.

Extended Data Fig. 5 Hierarchical Clustering of Mutational Clusters CCFs and Copy Number Alterations define concordant tumor lineages.

Top: Hierarchically clustered heatmap of inferred cancer cell fractions (CCFs) for each mutation cluster (columns) for each tumor (rows), demonstrating 7 different lineages. Bottom: Hierarchically clustered heatmap of large copy number alterations (columns) for each tumor (rows), demonstrating concordance with lineages derived from mutational clusters. Complete allelic deletions are dark blue, and copy number gains and losses are light blue.

Extended Data Fig. 6 Aneuploidy in Genome-Doubled vs. non-Genome-Doubled tumors.

Aneuploidy here is defined as the proportion of the genome with copy number gain or loss (compared to the ‘baseline’ allelic copy number, which is 1 for non-genome doubled tumors and 2 for genome doubled tumors). a, Allelic copy number ratios for a representative non-genome doubled tumor (T13 from lineage 1, upper), and a genome-doubled tumor (R1, the jejunal metastasis from lineage 3, lower). The x axis is the genome (chromosomes in increasing number), and y represents the relative inferred copy number at that genomic location. b, A different representation of the inferred allelic copy from T13 and R1 demonstrating increased aneuploidy in the genome-doubled tumor. c, The genome doubled tumors (n=19) had evidence of chromosomal instability, with higher proportion of genome with aneuploidy (two-sided Mann-Whitney p=2.2e-07, Methods) compared to non-genome doubled tumors (n=18) (upper panel). Late resistant tumors (D1500+) had the higher aneuploidy compared to all other tumors, (lower panel). Boxplots: box limits indicate the IQR (25th to 75th percentiles), with a center line indicating the median. Whiskers show the value ranges up to 1.5 × IQR above the 75th or below the 25th percentiles, with outliers beyond those ranges shown as individual points.

Extended Data Fig. 7 Tumor Immune Microenvironment.

a, Tumors from the patient (n=20) had lower overall immune signature score compared to a large cohort of PD-1 treated melanoma patients (n=121) (MWW nominal two-sided p = 0.036);. b, overall Immune signature scores in tumors biopsied within the first 120 days after immunotherapy initiation decreased after initiation of immune checkpoint blockade (linear regression p = 0.020). c, T cell signature scores in the same tumors decrease after initiation of immune checkpoint blockade (linear regression p = 0.008). d, All immune cell signature scores in the same tumors and their association with time after treatment. Negative coefficients are associated with a decrease in score with time after treatment. Boxplots: box limits indicate the IQR (25th to 75th percentiles), with a center line indicating the median. Whiskers show the value ranges up to 1.5 × IQR above the 75th or below the 25th percentiles, with outliers beyond those ranges shown as individual points.

Extended Data Fig. 8 Quantification of Selected Immune and Tumor Populations from CyCIF.

Selected immune cell and NGFR-high subset proportions over time by spatial compartment. (a) CD4 + Treg cells; (b) NGFR-high tumor cells. Error bars represent standard error of the mean (S.E.M.). Sample numbers for each days are: 1(day -62), 1(day -22), 4(day 4), 4(day 39), 2(day 62), 3(day 76), 3(day 92), 1(day 109), 1(day 1028), 1(day 1849), 1(day 1862), 10(day 2065).

Extended Data Fig. 9 Gaussian Mixture Modelling of CyCIF data.

a, Heatmap demonstrating clusters of cells from Gaussian Mixture Model clustering characterized by a range of CyCIF quantitative fluorescence (Methods) from the 19 tumors in Batch 1. b, Heatmap demonstrating clusters of cells from Gaussian Mixture Model clustering characterized by a range of CyCIF quantitative fluorescence (Methods) from the pre- and post-TLR9 + antiPD1 therapy tumors (Batch 2). (Top) There is a distinct NGFR-Hi tumor cell cluster, which is high in PD-L1 protein expression, a MITF-Hi/NGFR-lo tumor cell cluster, and an immune cell cluster. Several non-specific (that is non-NGFR-Hi, non-MITF-Hi) tumor cell clusters are also seen. (Bottom) There is a strong association between NGFR and PD-L1 expression among S100 + gated tumor cells (Pearson correlation coefficient r = 0.66 and p-value = 0, calculated by the default function in MATLAB).

Extended Data Fig. 10 Expression of class I and class II MHC and of clonal ancestral neoantigens in lineage 3 and over time.

a, MHC-I and –II scores were generated from bulk RNAseq and compared between Lineage 3 tumors (n=3) and other tumors (n=17). Scores for each sample were calculated using an averaged standardized z-score of 6 MHC-I genes (HLA-A, HLA-B, HLA-C, B2M, TAP1, TAP2) and 13 MHC-II genes (HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DPA1, HLA-DPB1, HLA-DQA1, HLA-DQA2, HLA-DQB1, HLA-DQB2, HLA-DRA, HLA-DRB1, HLA-DRB5). (Left) Lineage 3 has lower MHC-I score compared to other tumors (t-test p = 0.01). (Right) Lineage 3 tumors do not have a statistically significant difference in MHC-II score compared to other tumors (t-test p = 0.13). b, Expression of clonal ancestral neoantigens in lineage 3 and over time. Neoantigens were inferred using NetMHCPan with inputs of the patient’s HLA and mutations. 174 genes with clonal ancestral mutations that coded for neoantigens were identified, and their RNAseq expression (TPM) in each tumor calculated. Overall expression, Chr15 neoantigen expression (that is the expression of the 3 genes with clonal ancestral mutations lost with LOH of Chr15 in Lineage 3 tumors), and the proportion of the overall neoantigen expression that Chr15 neoantigen genes represented were calculated. Left: Lineage 3 vs other tumors. Overall expression was not different, but there was a trend towards lower expression and proportion of expression of Chr15 neoantigen genes in Lineage 3 tumors. Right: Expression over time in the on-treatment time period (D27-D109). Overall neoantigen gene expression was not different by time, but Chr15 neoantigen gene expression and the proportion of Chr15 neoantigen gene expression trended towards decreasing with time. Boxplots: box limits indicate the IQR (25th to 75th percentiles), with a center line indicating the median. Whiskers show the value ranges up to 1.5 × IQR above the 75th or below the 25th percentiles, with outliers beyond those ranges shown as individual points.

Supplementary information

Supplementary Information

Supplementary Figs. 1–9.

Reporting Summary

Supplementary Tables

Supplementary Tables 1–4

Supplementary Data 1

MAF (mutation annotation format) of called mutations per sample.

Supplementary Data 2

MAF (mutation annotation format) of union of all called mutations across samples with cancer cell fractions per sample used for clustering and phylogenetic analysis.

Supplementary Data 3

TPM (transcripts per kilobase million) normalized transcriptomes of tumor samples.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, D., Lin, JR., Robitschek, E.J. et al. Evolution of delayed resistance to immunotherapy in a melanoma responder. Nat Med 27, 985–992 (2021). https://doi.org/10.1038/s41591-021-01331-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-021-01331-8

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